The relatively long-lived phagocytic cell of mammalian tissues that are derived from blood MONOCYTES. Main types are PERITONEAL MACROPHAGES; ALVEOLAR MACROPHAGES; HISTIOCYTES; KUPFFER CELLS of the liver; and OSTEOCLASTS. They may further differentiate within chronic inflammatory lesions to EPITHELIOID CELLS or may fuse to form FOREIGN BODY GIANT CELLS or LANGHANS GIANT CELLS. (from The Dictionary of Cell Biology, Lackie and Dow, 3rd ed.)
The process of altering the morphology and functional activity of macrophages so that they become avidly phagocytic. It is initiated by lymphokines, such as the macrophage activation factor (MAF) and the macrophage migration-inhibitory factor (MMIF), immune complexes, C3b, and various peptides, polysaccharides, and immunologic adjuvants.
Mononuclear phagocytes derived from bone marrow precursors but resident in the peritoneum.
Round, granular, mononuclear phagocytes found in the alveoli of the lungs. They ingest small inhaled particles resulting in degradation and presentation of the antigen to immunocompetent cells.
The engulfing and degradation of microorganisms; other cells that are dead, dying, or pathogenic; and foreign particles by phagocytic cells (PHAGOCYTES).
A mononuclear phagocyte colony-stimulating factor (M-CSF) synthesized by mesenchymal cells. The compound stimulates the survival, proliferation, and differentiation of hematopoietic cells of the monocyte-macrophage series. M-CSF is a disulfide-bonded glycoprotein dimer with a MW of 70 kDa. It binds to a specific high affinity receptor (RECEPTOR, MACROPHAGE COLONY-STIMULATING FACTOR).
Cells propagated in vitro in special media conducive to their growth. Cultured cells are used to study developmental, morphologic, metabolic, physiologic, and genetic processes, among others.
Lipid-containing polysaccharides which are endotoxins and important group-specific antigens. They are often derived from the cell wall of gram-negative bacteria and induce immunoglobulin secretion. The lipopolysaccharide molecule consists of three parts: LIPID A, core polysaccharide, and O-specific chains (O ANTIGENS). When derived from Escherichia coli, lipopolysaccharides serve as polyclonal B-cell mitogens commonly used in laboratory immunology. (From Dorland, 28th ed)
Inbred C57BL mice are a strain of laboratory mice that have been produced by many generations of brother-sister matings, resulting in a high degree of genetic uniformity and homozygosity, making them widely used for biomedical research, including studies on genetics, immunology, cancer, and neuroscience.
Large, phagocytic mononuclear leukocytes produced in the vertebrate BONE MARROW and released into the BLOOD; contain a large, oval or somewhat indented nucleus surrounded by voluminous cytoplasm and numerous organelles.
Proteins released by sensitized LYMPHOCYTES and possibly other cells that inhibit the migration of MACROPHAGES away from the release site. The structure and chemical properties may vary with the species and type of releasing cell.
Established cell cultures that have the potential to propagate indefinitely.
Serum glycoprotein produced by activated MACROPHAGES and other mammalian MONONUCLEAR LEUKOCYTES. It has necrotizing activity against tumor cell lines and increases ability to reject tumor transplants. Also known as TNF-alpha, it is only 30% homologous to TNF-beta (LYMPHOTOXIN), but they share TNF RECEPTORS.
Inbred BALB/c mice are a strain of laboratory mice that have been selectively bred to be genetically identical to each other, making them useful for scientific research and experiments due to their consistent genetic background and predictable responses to various stimuli or treatments.
Non-antibody proteins secreted by inflammatory leukocytes and some non-leukocytic cells, that act as intercellular mediators. They differ from classical hormones in that they are produced by a number of tissue or cell types rather than by specialized glands. They generally act locally in a paracrine or autocrine rather than endocrine manner.
Strains of mice in which certain GENES of their GENOMES have been disrupted, or "knocked-out". To produce knockouts, using RECOMBINANT DNA technology, the normal DNA sequence of the gene being studied is altered to prevent synthesis of a normal gene product. Cloned cells in which this DNA alteration is successful are then injected into mouse EMBRYOS to produce chimeric mice. The chimeric mice are then bred to yield a strain in which all the cells of the mouse contain the disrupted gene. Knockout mice are used as EXPERIMENTAL ANIMAL MODELS for diseases (DISEASE MODELS, ANIMAL) and to clarify the functions of the genes.
The space enclosed by the peritoneum. It is divided into two portions, the greater sac and the lesser sac or omental bursa, which lies behind the STOMACH. The two sacs are connected by the foramen of Winslow, or epiploic foramen.
Organic esters of thioglycolic acid (HS-CH2COOH).
Heparin-binding proteins that exhibit a number of inflammatory and immunoregulatory activities. Originally identified as secretory products of MACROPHAGES, these chemokines are produced by a variety of cell types including NEUTROPHILS; FIBROBLASTS; and EPITHELIAL CELLS. They likely play a significant role in respiratory tract defenses.
A pathological process characterized by injury or destruction of tissues caused by a variety of cytologic and chemical reactions. It is usually manifested by typical signs of pain, heat, redness, swelling, and loss of function.
Surface antigens expressed on myeloid cells of the granulocyte-monocyte-histiocyte series during differentiation. Analysis of their reactivity in normal and malignant myelomonocytic cells is useful in identifying and classifying human leukemias and lymphomas.
The serous fluid of ASCITES, the accumulation of fluids in the PERITONEAL CAVITY.
A receptor for MACROPHAGE COLONY-STIMULATING FACTOR encoded by the c-fms proto-oncogene (GENES, FMS). It contains an intrinsic protein-tyrosine kinase activity. When activated the receptor undergoes autophosphorylation, phosphorylation of down-stream signaling molecules and rapid down-regulation.
RNA sequences that serve as templates for protein synthesis. Bacterial mRNAs are generally primary transcripts in that they do not require post-transcriptional processing. Eukaryotic mRNA is synthesized in the nucleus and must be exported to the cytoplasm for translation. Most eukaryotic mRNAs have a sequence of polyadenylic acid at the 3' end, referred to as the poly(A) tail. The function of this tail is not known for certain, but it may play a role in the export of mature mRNA from the nucleus as well as in helping stabilize some mRNA molecules by retarding their degradation in the cytoplasm.
The major interferon produced by mitogenically or antigenically stimulated LYMPHOCYTES. It is structurally different from TYPE I INTERFERON and its major activity is immunoregulation. It has been implicated in the expression of CLASS II HISTOCOMPATIBILITY ANTIGENS in cells that do not normally produce them, leading to AUTOIMMUNE DISEASES.
A diphosphonate which affects calcium metabolism. It inhibits bone resorption and soft tissue calcification.
Inbred C3H mice are a strain of laboratory mice that have been selectively bred to maintain a high degree of genetic uniformity and share specific genetic characteristics, including susceptibility to certain diseases, which makes them valuable for biomedical research purposes.
A family of scavenger receptors that mediate the influx of LIPIDS into MACROPHAGES and are involved in FOAM CELL formation.
Membrane-bound cytoplasmic vesicles formed by invagination of phagocytized material. They fuse with lysosomes to form phagolysosomes in which the hydrolytic enzymes of the lysosome digest the phagocytized material.
Any of the processes by which nuclear, cytoplasmic, or intercellular factors influence the differential control (induction or repression) of gene action at the level of transcription or translation.
A free radical gas produced endogenously by a variety of mammalian cells, synthesized from ARGININE by NITRIC OXIDE SYNTHASE. Nitric oxide is one of the ENDOTHELIUM-DEPENDENT RELAXING FACTORS released by the vascular endothelium and mediates VASODILATION. It also inhibits platelet aggregation, induces disaggregation of aggregated platelets, and inhibits platelet adhesion to the vascular endothelium. Nitric oxide activates cytosolic GUANYLATE CYCLASE and thus elevates intracellular levels of CYCLIC GMP.
Small polyhedral outpouchings along the walls of the alveolar sacs, alveolar ducts and terminal bronchioles through the walls of which gas exchange between alveolar air and pulmonary capillary blood takes place.
A CALCIUM-independent subtype of nitric oxide synthase that may play a role in immune function. It is an inducible enzyme whose expression is transcriptionally regulated by a variety of CYTOKINES.
The intracellular transfer of information (biological activation/inhibition) through a signal pathway. In each signal transduction system, an activation/inhibition signal from a biologically active molecule (hormone, neurotransmitter) is mediated via the coupling of a receptor/enzyme to a second messenger system or to an ion channel. Signal transduction plays an important role in activating cellular functions, cell differentiation, and cell proliferation. Examples of signal transduction systems are the GAMMA-AMINOBUTYRIC ACID-postsynaptic receptor-calcium ion channel system, the receptor-mediated T-cell activation pathway, and the receptor-mediated activation of phospholipases. Those coupled to membrane depolarization or intracellular release of calcium include the receptor-mediated activation of cytotoxic functions in granulocytes and the synaptic potentiation of protein kinase activation. Some signal transduction pathways may be part of larger signal transduction pathways; for example, protein kinase activation is part of the platelet activation signal pathway.
An encapsulated lymphatic organ through which venous blood filters.
Factors secreted by stimulated lymphocytes that prime macrophages to become nonspecifically cytotoxic to tumors. They also modulate the expression of macrophage cell surface Ia antigens. One MAF is INTERFERON-GAMMA. Other factors antigenically distinct from IFN-gamma have also been identified.
Zymosan is a polysaccharide derived from the cell walls of Saccharomyces cerevisiae, commonly used in research as an immunostimulant to induce inflammation and study phagocytosis, complement activation, and oxidative burst in neutrophils and macrophages.
Cells contained in the bone marrow including fat cells (see ADIPOCYTES); STROMAL CELLS; MEGAKARYOCYTES; and the immediate precursors of most blood cells.
A class of lipoproteins of small size (18-25 nm) and light (1.019-1.063 g/ml) particles with a core composed mainly of CHOLESTEROL ESTERS and smaller amounts of TRIGLYCERIDES. The surface monolayer consists mostly of PHOSPHOLIPIDS, a single copy of APOLIPOPROTEIN B-100, and free cholesterol molecules. The main LDL function is to transport cholesterol and cholesterol esters to extrahepatic tissues.
A subclass of lectins that are specific for CARBOHYDRATES that contain MANNOSE.
A chemokine that is a chemoattractant for MONOCYTES and may also cause cellular activation of specific functions related to host defense. It is produced by LEUKOCYTES of both monocyte and lymphocyte lineage and by FIBROBLASTS during tissue injury. It has specificity for CCR2 RECEPTORS.
Progressive restriction of the developmental potential and increasing specialization of function that leads to the formation of specialized cells, tissues, and organs.
Either of the pair of organs occupying the cavity of the thorax that effect the aeration of the blood.
A large group of structurally diverse cell surface receptors that mediate endocytic uptake of modified LIPOPROTEINS. Scavenger receptors are expressed by MYELOID CELLS and some ENDOTHELIAL CELLS, and were originally characterized based on their ability to bind acetylated LOW-DENSITY LIPOPROTEINS. They can also bind a variety of other polyanionic ligand. Certain scavenger receptors can internalize micro-organisms as well as apoptotic cells.
Technique using an instrument system for making, processing, and displaying one or more measurements on individual cells obtained from a cell suspension. Cells are usually stained with one or more fluorescent dyes specific to cell components of interest, e.g., DNA, and fluorescence of each cell is measured as it rapidly transverses the excitation beam (laser or mercury arc lamp). Fluorescence provides a quantitative measure of various biochemical and biophysical properties of the cell, as well as a basis for cell sorting. Other measurable optical parameters include light absorption and light scattering, the latter being applicable to the measurement of cell size, shape, density, granularity, and stain uptake.
Ubiquitous, inducible, nuclear transcriptional activator that binds to enhancer elements in many different cell types and is activated by pathogenic stimuli. The NF-kappa B complex is a heterodimer composed of two DNA-binding subunits: NF-kappa B1 and relA.
A pattern recognition receptor that interacts with LYMPHOCYTE ANTIGEN 96 and LIPOPOLYSACCHARIDES. It mediates cellular responses to GRAM-NEGATIVE BACTERIA.
Cell surface proteins that bind signalling molecules external to the cell with high affinity and convert this extracellular event into one or more intracellular signals that alter the behavior of the target cell (From Alberts, Molecular Biology of the Cell, 2nd ed, pp693-5). Cell surface receptors, unlike enzymes, do not chemically alter their ligands.
A thickening and loss of elasticity of the walls of ARTERIES that occurs with formation of ATHEROSCLEROTIC PLAQUES within the ARTERIAL INTIMA.
Naturally occurring or experimentally induced animal diseases with pathological processes sufficiently similar to those of human diseases. They are used as study models for human diseases.
Histochemical localization of immunoreactive substances using labeled antibodies as reagents.
The capacity of a normal organism to remain unaffected by microorganisms and their toxins. It results from the presence of naturally occurring ANTI-INFECTIVE AGENTS, constitutional factors such as BODY TEMPERATURE and immediate acting immune cells such as NATURAL KILLER CELLS.
Granular leukocytes having a nucleus with three to five lobes connected by slender threads of chromatin, and cytoplasm containing fine inconspicuous granules and stainable by neutral dyes.
A cytokine that stimulates the growth and differentiation of B-LYMPHOCYTES and is also a growth factor for HYBRIDOMAS and plasmacytomas. It is produced by many different cells including T-LYMPHOCYTES; MONOCYTES; and FIBROBLASTS.
Cell surface molecules on cells of the immune system that specifically bind surface molecules or messenger molecules and trigger changes in the behavior of cells. Although these receptors were first identified in the immune system, many have important functions elsewhere.
A soluble factor produced by MONOCYTES; MACROPHAGES, and other cells which activates T-lymphocytes and potentiates their response to mitogens or antigens. Interleukin-1 is a general term refers to either of the two distinct proteins, INTERLEUKIN-1ALPHA and INTERLEUKIN-1BETA. The biological effects of IL-1 include the ability to replace macrophage requirements for T-cell activation.
The endogenous compounds that mediate inflammation (AUTACOIDS) and related exogenous compounds including the synthetic prostaglandins (PROSTAGLANDINS, SYNTHETIC).
One of the mechanisms by which CELL DEATH occurs (compare with NECROSIS and AUTOPHAGOCYTOSIS). Apoptosis is the mechanism responsible for the physiological deletion of cells and appears to be intrinsically programmed. It is characterized by distinctive morphologic changes in the nucleus and cytoplasm, chromatin cleavage at regularly spaced sites, and the endonucleolytic cleavage of genomic DNA; (DNA FRAGMENTATION); at internucleosomal sites. This mode of cell death serves as a balance to mitosis in regulating the size of animal tissues and in mediating pathologic processes associated with tumor growth.
An acidic glycoprotein of MW 23 kDa with internal disulfide bonds. The protein is produced in response to a number of inflammatory mediators by mesenchymal cells present in the hemopoietic environment and at peripheral sites of inflammation. GM-CSF is able to stimulate the production of neutrophilic granulocytes, macrophages, and mixed granulocyte-macrophage colonies from bone marrow cells and can stimulate the formation of eosinophil colonies from fetal liver progenitor cells. GM-CSF can also stimulate some functional activities in mature granulocytes and macrophages.
Lymphocytes responsible for cell-mediated immunity. Two types have been identified - cytotoxic (T-LYMPHOCYTES, CYTOTOXIC) and helper T-lymphocytes (T-LYMPHOCYTES, HELPER-INDUCER). They are formed when lymphocytes circulate through the THYMUS GLAND and differentiate to thymocytes. When exposed to an antigen, they divide rapidly and produce large numbers of new T cells sensitized to that antigen.
Differentiation antigens residing on mammalian leukocytes. CD stands for cluster of differentiation, which refers to groups of monoclonal antibodies that show similar reactivity with certain subpopulations of antigens of a particular lineage or differentiation stage. The subpopulations of antigens are also known by the same CD designation.
A cytokine produced by a variety of cell types, including T-LYMPHOCYTES; MONOCYTES; DENDRITIC CELLS; and EPITHELIAL CELLS that exerts a variety of effects on immunoregulation and INFLAMMATION. Interleukin-10 combines with itself to form a homodimeric molecule that is the biologically active form of the protein.
Elements of limited time intervals, contributing to particular results or situations.
Genetically identical individuals developed from brother and sister matings which have been carried out for twenty or more generations, or by parent x offspring matings carried out with certain restrictions. All animals within an inbred strain trace back to a common ancestor in the twentieth generation.
A variation of the PCR technique in which cDNA is made from RNA via reverse transcription. The resultant cDNA is then amplified using standard PCR protocols.
Washing liquid obtained from irrigation of the lung, including the BRONCHI and the PULMONARY ALVEOLI. It is generally used to assess biochemical, inflammatory, or infection status of the lung.
Class of pro-inflammatory cytokines that have the ability to attract and activate leukocytes. They can be divided into at least three structural branches: C; (CHEMOKINES, C); CC; (CHEMOKINES, CC); and CXC; (CHEMOKINES, CXC); according to variations in a shared cysteine motif.
A class of morphologically heterogeneous cytoplasmic particles in animal and plant tissues characterized by their content of hydrolytic enzymes and the structure-linked latency of these enzymes. The intracellular functions of lysosomes depend on their lytic potential. The single unit membrane of the lysosome acts as a barrier between the enzymes enclosed in the lysosome and the external substrate. The activity of the enzymes contained in lysosomes is limited or nil unless the vesicle in which they are enclosed is ruptured. Such rupture is supposed to be under metabolic (hormonal) control. (From Rieger et al., Glossary of Genetics: Classical and Molecular, 5th ed)
A positive regulatory effect on physiological processes at the molecular, cellular, or systemic level. At the molecular level, the major regulatory sites include membrane receptors, genes (GENE EXPRESSION REGULATION), mRNAs (RNA, MESSENGER), and proteins.
Cells that can carry out the process of PHAGOCYTOSIS.
A CC chemokine with specificity for CCR5 RECEPTORS. It is a chemoattractant for NK CELLS; MONOCYTES and a variety of other immune cells. This chemokine is encoded by multiple genes.
A CC chemokine with specificity for CCR1 RECEPTORS and CCR5 RECEPTORS. It is a chemoattractant for NK CELLS; MONOCYTES; and a variety of other immune cells. This chemokine is encoded by multiple genes.
Proteins prepared by recombinant DNA technology.
The phenotypic manifestation of a gene or genes by the processes of GENETIC TRANSCRIPTION and GENETIC TRANSLATION.
A ureahydrolase that catalyzes the hydrolysis of arginine or canavanine to yield L-ornithine (ORNITHINE) and urea. Deficiency of this enzyme causes HYPERARGININEMIA. EC 3.5.3.1.
The principal sterol of all higher animals, distributed in body tissues, especially the brain and spinal cord, and in animal fats and oils.
Molecules found on the surface of some, but not all, B-lymphocytes, T-lymphocytes, and macrophages, which recognize and combine with the Fc (crystallizable) portion of immunoglobulin molecules.
The movement of cells from one location to another. Distinguish from CYTOKINESIS which is the process of dividing the CYTOPLASM of a cell.
A class of animal lectins that bind to carbohydrate in a calcium-dependent manner. They share a common carbohydrate-binding domain that is structurally distinct from other classes of lectins.
A pattern recognition receptor that forms heterodimers with other TOLL-LIKE RECEPTORS. It interacts with multiple ligands including PEPTIDOGLYCAN, bacterial LIPOPROTEINS, lipoarabinomannan, and a variety of PORINS.
An interleukin-1 subtype that is synthesized as an inactive membrane-bound pro-protein. Proteolytic processing of the precursor form by CASPASE 1 results in release of the active form of interleukin-1beta from the membrane.
The number of CELLS of a specific kind, usually measured per unit volume or area of sample.
The bovine variety of the tubercle bacillus. It is called also Mycobacterium tuberculosis var. bovis.
Soluble mediators of the immune response that are neither antibodies nor complement. They are produced largely, but not exclusively, by monocytes and macrophages.
Proteins that bind to particles and cells to increase susceptibility to PHAGOCYTOSIS, especially ANTIBODIES bound to EPITOPES that attach to FC RECEPTORS. COMPLEMENT C3B may also participate.
The engulfing of liquids by cells by a process of invagination and closure of the cell membrane to form fluid-filled vacuoles.
A species of gram-positive, aerobic bacteria that produces TUBERCULOSIS in humans, other primates, CATTLE; DOGS; and some other animals which have contact with humans. Growth tends to be in serpentine, cordlike masses in which the bacilli show a parallel orientation.
Glycoproteins found on the membrane or surface of cells.
Thickening and loss of elasticity of the walls of ARTERIES of all sizes. There are many forms classified by the types of lesions and arteries involved, such as ATHEROSCLEROSIS with fatty lesions in the ARTERIAL INTIMA of medium and large muscular arteries.
A class of protein components which can be found in several lipoproteins including HIGH-DENSITY LIPOPROTEINS; VERY-LOW-DENSITY LIPOPROTEINS; and CHYLOMICRONS. Synthesized in most organs, Apo E is important in the global transport of lipids and cholesterol throughout the body. Apo E is also a ligand for LDL receptors (RECEPTORS, LDL) that mediates the binding, internalization, and catabolism of lipoprotein particles in cells. There are several allelic isoforms (such as E2, E3, and E4). Deficiency or defects in Apo E are causes of HYPERLIPOPROTEINEMIA TYPE III.
Specific molecular sites on the surface of various cells, including B-lymphocytes and macrophages, that combine with IMMUNOGLOBULIN Gs. Three subclasses exist: Fc gamma RI (the CD64 antigen, a low affinity receptor), Fc gamma RII (the CD32 antigen, a high affinity receptor), and Fc gamma RIII (the CD16 antigen, a low affinity receptor).
Descriptions of specific amino acid, carbohydrate, or nucleotide sequences which have appeared in the published literature and/or are deposited in and maintained by databanks such as GENBANK, European Molecular Biology Laboratory (EMBL), National Biomedical Research Foundation (NBRF), or other sequence repositories.
The most common and most biologically active of the mammalian prostaglandins. It exhibits most biological activities characteristic of prostaglandins and has been used extensively as an oxytocic agent. The compound also displays a protective effect on the intestinal mucosa.
The span of viability of a cell characterized by the capacity to perform certain functions such as metabolism, growth, reproduction, some form of responsiveness, and adaptability.
An NADPH-dependent enzyme that catalyzes the conversion of L-ARGININE and OXYGEN to produce CITRULLINE and NITRIC OXIDE.
Specialized cells of the hematopoietic system that have branch-like extensions. They are found throughout the lymphatic system, and in non-lymphoid tissues such as SKIN and the epithelia of the intestinal, respiratory, and reproductive tracts. They trap and process ANTIGENS, and present them to T-CELLS, thereby stimulating CELL-MEDIATED IMMUNITY. They are different from the non-hematopoietic FOLLICULAR DENDRITIC CELLS, which have a similar morphology and immune system function, but with respect to humoral immunity (ANTIBODY PRODUCTION).
A technique of culturing mixed cell types in vitro to allow their synergistic or antagonistic interactions, such as on CELL DIFFERENTIATION or APOPTOSIS. Coculture can be of different types of cells, tissues, or organs from normal or disease states.
White blood cells formed in the body's lymphoid tissue. The nucleus is round or ovoid with coarse, irregularly clumped chromatin while the cytoplasm is typically pale blue with azurophilic (if any) granules. Most lymphocytes can be classified as either T or B (with subpopulations of each), or NATURAL KILLER CELLS.
A family of pattern recognition receptors characterized by an extracellular leucine-rich domain and a cytoplasmic domain that share homology with the INTERLEUKIN 1 RECEPTOR and the DROSOPHILA toll protein. Following pathogen recognition, toll-like receptors recruit and activate a variety of SIGNAL TRANSDUCING ADAPTOR PROTEINS.
Identification of proteins or peptides that have been electrophoretically separated by blot transferring from the electrophoresis gel to strips of nitrocellulose paper, followed by labeling with antibody probes.
A superfamily of large integral ATP-binding cassette membrane proteins whose expression pattern is consistent with a role in lipid (cholesterol) efflux. It is implicated in TANGIER DISEASE characterized by accumulation of cholesteryl ester in various tissues.
A CXC chemokine that is synthesized by activated MONOCYTES and NEUTROPHILS. It has specificity for CXCR2 RECEPTORS.
Laboratory mice that have been produced from a genetically manipulated EGG or EMBRYO, MAMMALIAN.
Microscopy using an electron beam, instead of light, to visualize the sample, thereby allowing much greater magnification. The interactions of ELECTRONS with specimens are used to provide information about the fine structure of that specimen. In TRANSMISSION ELECTRON MICROSCOPY the reactions of the electrons that are transmitted through the specimen are imaged. In SCANNING ELECTRON MICROSCOPY an electron beam falls at a non-normal angle on the specimen and the image is derived from the reactions occurring above the plane of the specimen.
Highly reactive compounds produced when oxygen is reduced by a single electron. In biological systems, they may be generated during the normal catalytic function of a number of enzymes and during the oxidation of hemoglobin to METHEMOGLOBIN. In living organisms, SUPEROXIDE DISMUTASE protects the cell from the deleterious effects of superoxides.
An adhesion-promoting leukocyte surface membrane heterodimer. The alpha subunit consists of the CD11b ANTIGEN and the beta subunit the CD18 ANTIGEN. The antigen, which is an integrin, functions both as a receptor for complement 3 and in cell-cell and cell-substrate adhesive interactions.
A heterodimeric cytokine that plays a role in innate and adaptive immune responses. Interleukin-12 is a 70 kDa protein that is composed of covalently linked 40 kDa and 35 kDa subunits. It is produced by DENDRITIC CELLS; MACROPHAGES and a variety of other immune cells and plays a role in the stimulation of INTERFERON-GAMMA production by T-LYMPHOCYTES and NATURAL KILLER CELLS.
A species of gram-positive, rod-shaped bacteria widely distributed in nature. It has been isolated from sewage, soil, silage, and from feces of healthy animals and man. Infection with this bacterium leads to encephalitis, meningitis, endocarditis, and abortion.
Specialized phagocytic cells of the MONONUCLEAR PHAGOCYTE SYSTEM found on the luminal surface of the hepatic sinusoids. They filter bacteria and small foreign proteins out of the blood, and dispose of worn out red blood cells.
Antibodies produced by a single clone of cells.
The relationship between the dose of an administered drug and the response of the organism to the drug.
A species of gram-negative, aerobic bacteria that is the causative agent of LEGIONNAIRES' DISEASE. It has been isolated from numerous environmental sites as well as from human lung tissue, respiratory secretions, and blood.
A secreted matrix metalloproteinase which is highly expressed by MACROPHAGES where it may play a role in INFLAMMATION and WOUND HEALING.
A bacterium causing tuberculosis in domestic fowl and other birds. In pigs, it may cause localized and sometimes disseminated disease. The organism occurs occasionally in sheep and cattle. It should be distinguished from the M. avium complex, which infects primarily humans.
Culture media containing biologically active components obtained from previously cultured cells or tissues that have released into the media substances affecting certain cell functions (e.g., growth, lysis).
A CD antigen that contains a conserved I domain which is involved in ligand binding. When combined with CD18 the two subunits form MACROPHAGE-1 ANTIGEN.
Adherence of cells to surfaces or to other cells.
A negative regulatory effect on physiological processes at the molecular, cellular, or systemic level. At the molecular level, the major regulatory sites include membrane receptors, genes (GENE EXPRESSION REGULATION), mRNAs (RNA, MESSENGER), and proteins.
A human cell line established from a diffuse histiocytic lymphoma (HISTIOCYTIC LYMPHOMA, DIFFUSE) and displaying many monocytic characteristics. It serves as an in vitro model for MONOCYTE and MACROPHAGE differentiation.
The species Oryctolagus cuniculus, in the family Leporidae, order LAGOMORPHA. Rabbits are born in burrows, furless, and with eyes and ears closed. In contrast with HARES, rabbits have 22 chromosome pairs.
An immunoassay utilizing an antibody labeled with an enzyme marker such as horseradish peroxidase. While either the enzyme or the antibody is bound to an immunosorbent substrate, they both retain their biologic activity; the change in enzyme activity as a result of the enzyme-antibody-antigen reaction is proportional to the concentration of the antigen and can be measured spectrophotometrically or with the naked eye. Many variations of the method have been developed.
Leukocyte differentiation antigens and major platelet membrane glycoproteins present on MONOCYTES; ENDOTHELIAL CELLS; PLATELETS; and mammary EPITHELIAL CELLS. They play major roles in CELL ADHESION; SIGNAL TRANSDUCTION; and regulation of angiogenesis. CD36 is a receptor for THROMBOSPONDINS and can act as a scavenger receptor that recognizes and transports oxidized LIPOPROTEINS and FATTY ACIDS.
Proteins which are found in membranes including cellular and intracellular membranes. They consist of two types, peripheral and integral proteins. They include most membrane-associated enzymes, antigenic proteins, transport proteins, and drug, hormone, and lectin receptors.
The process of intracellular viral multiplication, consisting of the synthesis of PROTEINS; NUCLEIC ACIDS; and sometimes LIPIDS, and their assembly into a new infectious particle.
The degree of pathogenicity within a group or species of microorganisms or viruses as indicated by case fatality rates and/or the ability of the organism to invade the tissues of the host. The pathogenic capacity of an organism is determined by its VIRULENCE FACTORS.
The outward appearance of the individual. It is the product of interactions between genes, and between the GENOTYPE and the environment.
The sequence of PURINES and PYRIMIDINES in nucleic acids and polynucleotides. It is also called nucleotide sequence.
The rate dynamics in chemical or physical systems.
The phenomenon of target cell destruction by immunologically active effector cells. It may be brought about directly by sensitized T-lymphocytes or by lymphoid or myeloid "killer" cells, or it may be mediated by cytotoxic antibody, cytotoxic factor released by lymphoid cells, or complement.
The third type of glial cell, along with astrocytes and oligodendrocytes (which together form the macroglia). Microglia vary in appearance depending on developmental stage, functional state, and anatomical location; subtype terms include ramified, perivascular, ameboid, resting, and activated. Microglia clearly are capable of phagocytosis and play an important role in a wide spectrum of neuropathologies. They have also been suggested to act in several other roles including in secretion (e.g., of cytokines and neural growth factors), in immunological processing (e.g., antigen presentation), and in central nervous system development and remodeling.
A relatively small nodular inflammatory lesion containing grouped mononuclear phagocytes, caused by infectious and noninfectious agents.
Large, transmembrane, non-covalently linked glycoproteins (alpha and beta). Both chains can be polymorphic although there is more structural variation in the beta chains. The class II antigens in humans are called HLA-D ANTIGENS and are coded by a gene on chromosome 6. In mice, two genes named IA and IE on chromosome 17 code for the H-2 antigens. The antigens are found on B-lymphocytes, macrophages, epidermal cells, and sperm and are thought to mediate the competence of and cellular cooperation in the immune response. The term IA antigens used to refer only to the proteins encoded by the IA genes in the mouse, but is now used as a generic term for any class II histocompatibility antigen.
Assays that measure the rate of migration of MACROPHAGES. They may involve the use hollow plastic chamber, sealed at one end with a porous membrane and suspended over a larger well which may contain CHEMOTACTIC FACTORS. The migration of cell through the pores to the other side of the membrane is measured.
The fission of a CELL. It includes CYTOKINESIS, when the CYTOPLASM of a cell is divided, and CELL NUCLEUS DIVISION.
Manifestations of the immune response which are mediated by antigen-sensitized T-lymphocytes via lymphokines or direct cytotoxicity. This takes place in the absence of circulating antibody or where antibody plays a subordinate role.
A sialic acid binding lectin that was originally identified as an adhesion molecule for inflammatory MACROPHAGES and activated MONOCYTES. This protein is the largest known siglec subtype and contains 16 immunoglobulin C2-set domains. It plays a role in cell to cell interactions and interactions with BACTERIA.
An intracellular signaling adaptor protein that plays a role in TOLL-LIKE RECEPTOR and INTERLEUKIN 1 RECEPTORS signal transduction. It forms a signaling complex with the activated cell surface receptors and members of the IRAK KINASES.
The movement of cells or organisms toward or away from a substance in response to its concentration gradient.
Fatty acid esters of cholesterol which constitute about two-thirds of the cholesterol in the plasma. The accumulation of cholesterol esters in the arterial intima is a characteristic feature of atherosclerosis.
Salts of nitrous acid or compounds containing the group NO2-. The inorganic nitrites of the type MNO2 (where M=metal) are all insoluble, except the alkali nitrites. The organic nitrites may be isomeric, but not identical with the corresponding nitro compounds. (Grant & Hackh's Chemical Dictionary, 5th ed)
The type species of LENTIVIRUS and the etiologic agent of AIDS. It is characterized by its cytopathic effect and affinity for the T4-lymphocyte.
Antigens expressed primarily on the membranes of living cells during sequential stages of maturation and differentiation. As immunologic markers they have high organ and tissue specificity and are useful as probes in studies of normal cell development as well as neoplastic transformation.
Substances that reduce or suppress INFLAMMATION.
A soluble factor produced by activated T-LYMPHOCYTES that induces the expression of MHC CLASS II GENES and FC RECEPTORS on B-LYMPHOCYTES and causes their proliferation and differentiation. It also acts on T-lymphocytes, MAST CELLS, and several other hematopoietic lineage cells.
A phorbol ester found in CROTON OIL with very effective tumor promoting activity. It stimulates the synthesis of both DNA and RNA.
A family of scavenger receptors that are predominately localized to CAVEOLAE of the PLASMA MEMBRANE and bind HIGH DENSITY LIPOPROTEINS.
Morphologic alteration of small B LYMPHOCYTES or T LYMPHOCYTES in culture into large blast-like cells able to synthesize DNA and RNA and to divide mitotically. It is induced by INTERLEUKINS; MITOGENS such as PHYTOHEMAGGLUTININS, and by specific ANTIGENS. It may also occur in vivo as in GRAFT REJECTION.
Phenomenon of cell-mediated immunity measured by in vitro inhibition of the migration or phagocytosis of antigen-stimulated LEUKOCYTES or MACROPHAGES. Specific CELL MIGRATION ASSAYS have been developed to estimate levels of migration inhibitory factors, immune reactivity against tumor-associated antigens, and immunosuppressive effects of infectious microorganisms.
Cell separation is the process of isolating and distinguishing specific cell types or individual cells from a heterogeneous mixture, often through the use of physical or biological techniques.
Inbred CBA mice are a strain of laboratory mice that have been selectively bred to be genetically identical and uniform, which makes them useful for scientific research, particularly in the areas of immunology and cancer.
White blood cells. These include granular leukocytes (BASOPHILS; EOSINOPHILS; and NEUTROPHILS) as well as non-granular leukocytes (LYMPHOCYTES and MONOCYTES).
A genus of flagellate protozoa comprising several species that are pathogenic for humans. Organisms of this genus have an amastigote and a promastigote stage in their life cycles. As a result of enzymatic studies this single genus has been divided into two subgenera: Leishmania leishmania and Leishmania viannia. Species within the Leishmania leishmania subgenus include: L. aethiopica, L. arabica, L. donovani, L. enrietti, L. gerbilli, L. hertigi, L. infantum, L. major, L. mexicana, and L. tropica. The following species are those that compose the Leishmania viannia subgenus: L. braziliensis, L. guyanensis, L. lainsoni, L. naiffi, and L. shawi.
Inbred ICR mice are a strain of albino laboratory mice that have been selectively bred for consistent genetic makeup and high reproductive performance, making them widely used in biomedical research for studies involving reproduction, toxicology, pharmacology, and carcinogenesis.
Chemical substances that attract or repel cells. The concept denotes especially those factors released as a result of tissue injury, microbial invasion, or immunologic activity, that attract LEUKOCYTES; MACROPHAGES; or other cells to the site of infection or insult.
Conversion of an inactive form of an enzyme to one possessing metabolic activity. It includes 1, activation by ions (activators); 2, activation by cofactors (coenzymes); and 3, conversion of an enzyme precursor (proenzyme or zymogen) to an active enzyme.
A bacteria isolated from normal skin, intestinal contents, wounds, blood, pus, and soft tissue abscesses. It is a common contaminant of clinical specimens, presumably from the skin of patients or attendants.
Transparent, tasteless crystals found in nature as agate, amethyst, chalcedony, cristobalite, flint, sand, QUARTZ, and tridymite. The compound is insoluble in water or acids except hydrofluoric acid.
Receptors on the plasma membrane of nonhepatic cells that specifically bind LDL. The receptors are localized in specialized regions called coated pits. Hypercholesteremia is caused by an allelic genetic defect of three types: 1, receptors do not bind to LDL; 2, there is reduced binding of LDL; and 3, there is normal binding but no internalization of LDL. In consequence, entry of cholesterol esters into the cell is impaired and the intracellular feedback by cholesterol on 3-hydroxy-3-methylglutaryl CoA reductase is lacking.
A large lobed glandular organ in the abdomen of vertebrates that is responsible for detoxification, metabolism, synthesis and storage of various substances.
The movement of leukocytes in response to a chemical concentration gradient or to products formed in an immunologic reaction.
Any of several ways in which living cells of an organism communicate with one another, whether by direct contact between cells or by means of chemical signals carried by neurotransmitter substances, hormones, and cyclic AMP.
A long pro-domain caspase that has specificity for the precursor form of INTERLEUKIN-1BETA. It plays a role in INFLAMMATION by catalytically converting the inactive forms of CYTOKINES such as interleukin-1beta to their active, secreted form. Caspase 1 is referred as interleukin-1beta converting enzyme and is frequently abbreviated ICE.
Toxins closely associated with the living cytoplasm or cell wall of certain microorganisms, which do not readily diffuse into the culture medium, but are released upon lysis of the cells.
An inducibly-expressed subtype of prostaglandin-endoperoxide synthase. It plays an important role in many cellular processes and INFLAMMATION. It is the target of COX2 INHIBITORS.
Molecules on the surface of some B-lymphocytes and macrophages, that recognize and combine with the C3b, C3d, C1q, and C4b components of complement.
An enzyme that catalyzes the conversion of an orthophosphoric monoester and water to an alcohol and orthophosphate. EC 3.1.3.2.
Cell surface proteins that bind lipoproteins with high affinity. Lipoprotein receptors in the liver and peripheral tissues mediate the regulation of plasma and cellular cholesterol metabolism and concentration. The receptors generally recognize the apolipoproteins of the lipoprotein complex, and binding is often a trigger for endocytosis.
CCR receptors with specificity for CHEMOKINE CCL2 and several other CCL2-related chemokines. They are expressed at high levels in T-LYMPHOCYTES; B-LYMPHOCYTES; MACROPHAGES; BASOPHILS; and NK CELLS.
Infections with bacteria of the genus LISTERIA.
Glycoproteins found in a subfraction of normal mammalian plasma and urine. They stimulate the proliferation of bone marrow cells in agar cultures and the formation of colonies of granulocytes and/or macrophages. The factors include INTERLEUKIN-3; (IL-3); GRANULOCYTE COLONY-STIMULATING FACTOR; (G-CSF); MACROPHAGE COLONY-STIMULATING FACTOR; (M-CSF); and GRANULOCYTE-MACROPHAGE COLONY-STIMULATING FACTOR; (GM-CSF).
Mice bearing mutant genes which are phenotypically expressed in the animals.
A milky, product excreted from the latex canals of a variety of plant species that contain cauotchouc. Latex is composed of 25-35% caoutchouc, 60-75% water, 2% protein, 2% resin, 1.5% sugar & 1% ash. RUBBER is made by the removal of water from latex.(From Concise Encyclopedia Biochemistry and Molecular Biology, 3rd ed). Hevein proteins are responsible for LATEX HYPERSENSITIVITY. Latexes are used as inert vehicles to carry antibodies or antigens in LATEX FIXATION TESTS.
Molecules or ions formed by the incomplete one-electron reduction of oxygen. These reactive oxygen intermediates include SINGLET OXYGEN; SUPEROXIDES; PEROXIDES; HYDROXYL RADICAL; and HYPOCHLOROUS ACID. They contribute to the microbicidal activity of PHAGOCYTES, regulation of signal transduction and gene expression, and the oxidative damage to NUCLEIC ACIDS; PROTEINS; and LIPIDS.
A large increase in oxygen uptake by neutrophils and most types of tissue macrophages through activation of an NADPH-cytochrome b-dependent oxidase that reduces oxygen to a superoxide. Individuals with an inherited defect in which the oxidase that reduces oxygen to superoxide is decreased or absent (GRANULOMATOUS DISEASE, CHRONIC) often die as a result of recurrent bacterial infections.
Compounds or agents that combine with an enzyme in such a manner as to prevent the normal substrate-enzyme combination and the catalytic reaction.
Transport proteins that carry specific substances in the blood or across cell membranes.
A parasitic hemoflagellate of the subgenus Leishmania leishmania that infects man and animals including rodents. The Leishmania mexicana complex causes both cutaneous (LEISHMANIASIS, CUTANEOUS) and diffuse cutaneous leishmaniasis (LEISHMANIASIS, DIFFUSE CUTANEOUS) and includes the subspecies amazonensis, garnhami, mexicana, pifanoi, and venezuelensis. L. m. mexicana causes chiclero ulcer, a form of cutaneous leishmaniasis (LEISHMANIASIS, CUTANEOUS) in the New World. The sandfly, Lutzomyia, appears to be the vector.
Artificial, single or multilaminar vesicles (made from lecithins or other lipids) that are used for the delivery of a variety of biological molecules or molecular complexes to cells, for example, drug delivery and gene transfer. They are also used to study membranes and membrane proteins.
Enzymes of the isomerase class that catalyze the oxidation of one part of a molecule with a corresponding reduction of another part of the same molecule. They include enzymes converting aldoses to ketoses (ALDOSE-KETOSE ISOMERASES), enzymes shifting a carbon-carbon double bond (CARBON-CARBON DOUBLE BOND ISOMERASES), and enzymes transposing S-S bonds (SULFUR-SULFUR BOND ISOMERASES). (From Enzyme Nomenclature, 1992) EC 5.3.
A mitogen-activated protein kinase subfamily that regulates a variety of cellular processes including CELL GROWTH PROCESSES; CELL DIFFERENTIATION; APOPTOSIS; and cellular responses to INFLAMMATION. The P38 MAP kinases are regulated by CYTOKINE RECEPTORS and can be activated in response to bacterial pathogens.
A parasitic hemoflagellate of the subgenus Leishmania leishmania that infects man and animals and causes visceral leishmaniasis (LEISHMANIASIS, VISCERAL). The sandfly genera Phlebotomus and Lutzomyia are the vectors.
A family of MEMBRANE TRANSPORT PROTEINS that require ATP hydrolysis for the transport of substrates across membranes. The protein family derives its name from the ATP-binding domain found on the protein.
A member of the CXC chemokine family that plays a role in the regulation of the acute inflammatory response. It is secreted by variety of cell types and induces CHEMOTAXIS of NEUTROPHILS and other inflammatory cells.
An increase in the rate of synthesis of an enzyme due to the presence of an inducer which acts to derepress the gene responsible for enzyme synthesis.
Test for tissue antigen using either a direct method, by conjugation of antibody with fluorescent dye (FLUORESCENT ANTIBODY TECHNIQUE, DIRECT) or an indirect method, by formation of antigen-antibody complex which is then labeled with fluorescein-conjugated anti-immunoglobulin antibody (FLUORESCENT ANTIBODY TECHNIQUE, INDIRECT). The tissue is then examined by fluorescence microscopy.
Mature LYMPHOCYTES and MONOCYTES transported by the blood to the body's extravascular space. They are morphologically distinguishable from mature granulocytic leukocytes by their large, non-lobed nuclei and lack of coarse, heavily stained cytoplasmic granules.
The introduction of a phosphoryl group into a compound through the formation of an ester bond between the compound and a phosphorus moiety.
A strong oxidizing agent used in aqueous solution as a ripening agent, bleach, and topical anti-infective. It is relatively unstable and solutions deteriorate over time unless stabilized by the addition of acetanilide or similar organic materials.
A CC-type chemokine that is a chemoattractant for EOSINOPHILS; MONOCYTES; and LYMPHOCYTES. It is a potent and selective eosinophil chemotaxin that is stored in and released from PLATELETS and activated T-LYMPHOCYTES. Chemokine CCL5 is specific for CCR1 RECEPTORS; CCR3 RECEPTORS; and CCR5 RECEPTORS. The acronym RANTES refers to Regulated on Activation, Normal T Expressed and Secreted.
Soluble protein factors generated by activated lymphocytes that affect other cells, primarily those involved in cellular immunity.
Substances that augment, stimulate, activate, potentiate, or modulate the immune response at either the cellular or humoral level. The classical agents (Freund's adjuvant, BCG, Corynebacterium parvum, et al.) contain bacterial antigens. Some are endogenous (e.g., histamine, interferon, transfer factor, tuftsin, interleukin-1). Their mode of action is either non-specific, resulting in increased immune responsiveness to a wide variety of antigens, or antigen-specific, i.e., affecting a restricted type of immune response to a narrow group of antigens. The therapeutic efficacy of many biological response modifiers is related to their antigen-specific immunoadjuvanticity.
The order of amino acids as they occur in a polypeptide chain. This is referred to as the primary structure of proteins. It is of fundamental importance in determining PROTEIN CONFORMATION.
A common name used for the genus Cavia. The most common species is Cavia porcellus which is the domesticated guinea pig used for pets and biomedical research.
The biosynthesis of RNA carried out on a template of DNA. The biosynthesis of DNA from an RNA template is called REVERSE TRANSCRIPTION.
Animals or humans raised in the absence of a particular disease-causing virus or other microorganism. Less frequently plants are cultivated pathogen-free.
A cytokine subunit that is a component of both interleukin-12 and interleukin-23. It binds to the INTERLEUKIN-12 SUBUNIT P35 via a disulfide bond to form interleukin-12 and to INTERLEUKIN-23 SUBUNIT P19 to form interleukin-23.
The uptake of naked or purified DNA by CELLS, usually meaning the process as it occurs in eukaryotic cells. It is analogous to bacterial transformation (TRANSFORMATION, BACTERIAL) and both are routinely employed in GENE TRANSFER TECHNIQUES.
Inbred DBA mice are a strain of laboratory mice that are genetically identical and share specific characteristics, including a high incidence of deafness, coat color (black and white), and susceptibility to certain diseases, which make them useful for research purposes in biomedical studies.
The major immunoglobulin isotype class in normal human serum. There are several isotype subclasses of IgG, for example, IgG1, IgG2A, and IgG2B.
A broad category of receptor-like proteins that may play a role in transcriptional-regulation in the CELL NUCLEUS. Many of these proteins are similar in structure to known NUCLEAR RECEPTORS but appear to lack a functional ligand-binding domain, while in other cases the specific ligands have yet to be identified.
Immunoglobulin molecules having a specific amino acid sequence by virtue of which they interact only with the ANTIGEN (or a very similar shape) that induced their synthesis in cells of the lymphoid series (especially PLASMA CELLS).
The soft tissue filling the cavities of bones. Bone marrow exists in two types, yellow and red. Yellow marrow is found in the large cavities of large bones and consists mostly of fat cells and a few primitive blood cells. Red marrow is a hematopoietic tissue and is the site of production of erythrocytes and granular leukocytes. Bone marrow is made up of a framework of connective tissue containing branching fibers with the frame being filled with marrow cells.

Hypersensitivity pneumonitis: experimental production in calves with antigens of Micropolyspora faeni. (1/32182)

Pneumonitis was induced in calves by exposure to aerosols of Micropolyspora faeni with or without prior sensitization of the animals by subcutaneous injection of antigen. The pneumonitis primarily involved centrolobular areas and was characterized by alveolar septal thickening and loss of air space by cellular infiltration. Vasculitis and focal haemorrhage occurred in certain individuals and haemoproteinaceous exudate appeared within septa and alveolar lumina. The pneumonitis was compared with human farmer's lung, pneumonitis of housed cattle and other experimental hypersensitivity pneumonitides.  (+info)

Cell-mediated immunity: dealing a direct blow to pathogens. (2/32182)

Cytotoxic T lymphocytes are essential for defence against viral infections. Recent data demonstrating direct killing of intracellular bacteria by granulysin, a protein released from the granules of cytotoxic T lymphocytes, emphasize the contribution of these lymphocytes to the control of tuberculosis.  (+info)

Chlamydial and human heat shock protein 60s activate human vascular endothelium, smooth muscle cells, and macrophages. (3/32182)

Both chlamydial and human heat shock protein 60s (HSP 60), which colocalize in human atheroma, may contribute to inflammation during atherogenesis. We tested the hypothesis that chlamydial or human HSP 60 activates human endothelial cells (ECs), smooth muscle cells (SMCs), and monocyte-derived macrophages. We examined the expression of adhesion molecules such as endothelial-leukocyte adhesion molecule-1 (E-selectin), intercellular adhesion molecule-1 (ICAM-1), and vascular cell adhesion molecule-1 (VCAM-1), and the production of the proinflammatory cytokine interleukin-6 (IL-6). We also tested whether either HSP 60 induces nuclear factor-kappaB (NF-kappaB), which contributes to the gene expression of these molecules. Either chlamydial or human HSP 60 induced E-selectin, ICAM-1, and VCAM-1 expression on ECs similar to levels induced by Escherichia coli lipopolysaccharide (LPS). Each HSP 60 also significantly induced IL-6 production by ECs, SMCs, and macrophages to an extent similar to that induced by E. coli LPS, as assessed by enzyme-linked immunosorbent assay (ELISA). In ECs, either HSP 60 triggered activation of NF-kappaB complexes containing p65 and p50 Rel proteins. Heat treatment abolished all these effects, but did not alter the ability of E. coli LPS to induce these functions. Chlamydial and human HSP 60s therefore activate human vascular cell functions relevant to atherogenesis and lesional complications. These findings help to elucidate the mechanisms by which a chronic asymptomatic chlamydial infection might contribute to the pathophysiology of atheroma.  (+info)

Anti-monocyte chemoattractant protein-1/monocyte chemotactic and activating factor antibody inhibits neointimal hyperplasia in injured rat carotid arteries. (4/32182)

Monocyte chemoattractant protein-1 (MCP-1)/monocyte chemotactic and activating factor (MCAF) has been suggested to promote atherogenesis. The effects of in vivo neutralization of MCP-1 in a rat model were examined in an effort to clarify the role of MCP-1 in the development of neointimal hyperplasia. Competitive polymerase chain reaction analysis revealed maximum MCP-1 mRNA expression at 4 hours after carotid arterial injury. Increased immunoreactivities of MCP-1 were also detected at 2 and 8 hours after injury. Either anti-MCP-1 antibody or nonimmunized goat IgG (10 mg/kg) was then administered every 12 hours to rats that had undergone carotid arterial injury. Treatment with 3 consecutive doses of anti-MCP-1 antibody within 24 hours (experiment 1) and every 12 hours for 5 days (experiment 2) significantly inhibited neointimal hyperplasia at day 14, resulting in a 27.8% reduction of the mean intima/media ratio (P<0.05) in experiment 1 and a 43.6% reduction (P<0.01) in experiment 2. This effect was still apparent at day 56 (55.6% inhibition; P<0.05). The number of vascular smooth muscle cells in the neointima at day 4 was significantly reduced by anti-MCP-1 treatment, demonstrating the important role of MCP-1 in early neointimal lesion formation. However, recombinant MCP-1 did not stimulate chemotaxis of vascular smooth muscle cells in an in vitro migration assay. These results suggest that MCP-1 promotes neointimal hyperplasia in early neointimal lesion formation and that neutralization of MCP-1 before, and immediately after, arterial injury may be effective in preventing restenosis after angioplasty. Further studies are needed to clarify the mechanism underlying the promotion of neointimal hyperplasia by MCP-1.  (+info)

Activated macrophages and microglia induce dopaminergic sprouting in the injured striatum and express brain-derived neurotrophic factor and glial cell line-derived neurotrophic factor. (5/32182)

Nigrostriatal dopaminergic neurons undergo sprouting around the margins of a striatal wound. The mechanism of this periwound sprouting has been unclear. In this study, we have examined the role played by the macrophage and microglial response that follows striatal injury. Macrophages and activated microglia quickly accumulate after injury and reach their greatest numbers in the first week. Subsequently, the number of both cell types declines rapidly in the first month and thereafter more slowly. Macrophage numbers eventually cease to decline, and a sizable group of these cells remains at the wound site and forms a long-term, highly activated resident population. This population of macrophages expresses increasing amounts of glial cell line-derived neurotrophic factor mRNA with time. Brain-derived neurotrophic factor mRNA is also expressed in and around the wound site. Production of this factor is by both activated microglia and, to a lesser extent, macrophages. The production of these potent dopaminergic neurotrophic factors occurs in a similar spatial distribution to sprouting dopaminergic fibers. Moreover, dopamine transporter-positive dopaminergic neurites can be seen growing toward and embracing hemosiderin-filled wound macrophages. The dopaminergic sprouting that accompanies striatal injury thus appears to result from neurotrophic factor secretion by activated macrophages and microglia at the wound site.  (+info)

Overexpression of CuZn superoxide dismutase protects RAW 264.7 macrophages against nitric oxide cytotoxicity. (6/32182)

Initiation of nitric oxide (NO.)-mediated apoptotic cell death in RAW 264.7 macrophages is associated with up-regulation of mitochondrial manganese superoxide dismutase (MnSOD; SOD2) and down-regulation of cytosolic copper zinc superoxide dismutase (CuZnSOD; SOD1) at their individual mRNA and protein levels. To evaluate the decreased CuZnSOD expression and the initiation of apoptosis we stably transfected macrophages to overexpress human CuZnSOD. Individual clones revealed a 2-fold increase in CuZnSOD activity. Expression of a functional and thus protective CuZnSOD was verified by attenuated superoxide (O2(.)-)-mediated apoptotic as well as necrotic cell death. In this study we showed that SOD-overexpressing macrophages (R-SOD1-12) were also protected against NO.-initiated programmed cell death. Protection was substantial towards NO. derived from exogenously added NO donors or when NO. was generated by inducible NO synthase activation, and was evident at the level of p53 accumulation, caspase activation and DNA fragmentation. Stimulation of parent and SOD-overexpressing cells with a combination of lipopolysaccharide and murine interferon gamma produced equivalent amounts of nitrite/nitrate, which ruled out attenuated inducible NO. synthase activity during protection. Because protection by a O2(.)--scavenging system during NO. -intoxication implies a role of NO. and O2(.)- in the progression of cell damage, we used uric acid to delineate the role of peroxynitrite during NO.-elicited apoptosis. The peroxynitrite scavenger uric acid left S-nitrosoglutathione or spermine-NO-elicited apoptosis unaltered, blocking only 3-morpholinosydnonimine-mediated cell death. As a result we exclude peroxynitrite from contributing, to any major extent, to NO. -mediated apoptosis. Therefore protection observed with CuZnSOD overexpression is unlikely to stem from interference with peroxynitrite formation and/or action. Unequivocally, the down-regulation of CuZnSOD is associated with NO. cytotoxicity, whereas CuZnSOD overexpression protects macrophages from apoptosis.  (+info)

Salmonella typhimurium and lipopolysaccharide stimulate extracellularly regulated kinase activation in macrophages by a mechanism involving phosphatidylinositol 3-kinase and phospholipase D as novel intermediates. (7/32182)

Activation of the extracellularly regulated kinase (ERK) pathway is part of the early biochemical events that follow lipopolysaccharide (LPS) treatment of macrophages or their infection by virulent and attenuated Salmonella strains. Phagocytosis as well as the secretion of invasion-associated proteins is dispensable for ERK activation by the pathogen. Furthermore, the pathways used by Salmonella and LPS to stimulate ERK are identical, suggesting that kinase activation might be solely mediated by LPS. Both stimuli activate ERK by a mechanism involving herbimycin-dependent tyrosine kinase(s) and phosphatidylinositol 3-kinase. Phospholipase D activation and stimulation of protein kinase C appear to be intermediates in this novel pathway of MEK/ERK activation.  (+info)

Non-serum-dependent chemotactic factors produced by Candida albicans stimulate chemotaxis by binding to the formyl peptide receptor on neutrophils and to an unknown receptor on macrophages. (8/32182)

Serum-free culture filtrates of six Candida species and Saccharomyces cerevisiae were found to contain chemoattractants for human polymorphonuclear leukocytes (PMNs) and a mouse macrophage-like cell line, J774. The chemotactic factors differed for the PMN and J774 cells, however, in terms of heat stability, kinetics of liberation by the yeast cells, and divalent cation requirements for production. The chemoattractant in Candida albicans culture filtrates appeared to act through the formyl peptide receptor (FPR) of PMNs, since it was found to induce chemotaxis of Chinese hamster ovary (CHO) cells that were expressing the human FPR but did not induce chemotaxis of wild-type CHO cells. The C. albicans culture filtrates also induced migration of PMNs across confluent monolayers of a human gastrointestinal epithelial cell line, T84; migration occurred in the basolateral-to-apical direction but not the reverse direction, unless the epithelial tight junctions were disrupted. J774 cells did not migrate toward the formylated peptide (fMet-Leu-Phe; fMLF), and chemotaxis toward the C. albicans culture filtrate was not inhibited by an FPR antagonist (t-butoxycarbonyl-Met-Leu-Phe), suggesting that a different receptor mediated J774 cell chemotaxis. In conclusion, we have identified a receptor by which a non-serum-dependent chemotactic factor (NSCF) produced by C. albicans induced chemotaxis of PMNs. Additionally, we have shown that NSCF was active across epithelial monolayers. These findings suggest that NSCFs produced by C. albicans and other yeast species may influence host-pathogen interactions at the gastrointestinal tract mucosal surface by inducing phagocytic-cell infiltration.  (+info)

Macrophages are a type of white blood cell that are an essential part of the immune system. They are large, specialized cells that engulf and destroy foreign substances, such as bacteria, viruses, parasites, and fungi, as well as damaged or dead cells. Macrophages are found throughout the body, including in the bloodstream, lymph nodes, spleen, liver, lungs, and connective tissues. They play a critical role in inflammation, immune response, and tissue repair and remodeling.

Macrophages originate from monocytes, which are a type of white blood cell produced in the bone marrow. When monocytes enter the tissues, they differentiate into macrophages, which have a larger size and more specialized functions than monocytes. Macrophages can change their shape and move through tissues to reach sites of infection or injury. They also produce cytokines, chemokines, and other signaling molecules that help coordinate the immune response and recruit other immune cells to the site of infection or injury.

Macrophages have a variety of surface receptors that allow them to recognize and respond to different types of foreign substances and signals from other cells. They can engulf and digest foreign particles, bacteria, and viruses through a process called phagocytosis. Macrophages also play a role in presenting antigens to T cells, which are another type of immune cell that helps coordinate the immune response.

Overall, macrophages are crucial for maintaining tissue homeostasis, defending against infection, and promoting wound healing and tissue repair. Dysregulation of macrophage function has been implicated in a variety of diseases, including cancer, autoimmune disorders, and chronic inflammatory conditions.

Macrophage activation is a process in which these immune cells become increasingly active and responsive to various stimuli, such as pathogens or inflammatory signals. This activation triggers a series of changes within the macrophages, allowing them to perform important functions like phagocytosis (ingesting and destroying foreign particles or microorganisms), antigen presentation (presenting microbial fragments to T-cells to stimulate an immune response), and production of cytokines and chemokines (signaling molecules that help coordinate the immune response).

There are two main types of macrophage activation: classical (or M1) activation and alternative (or M2) activation. Classical activation is typically induced by interferon-gamma (IFN-γ) and lipopolysaccharide (LPS), leading to a proinflammatory response, enhanced microbicidal activity, and the production of reactive oxygen and nitrogen species. Alternative activation, on the other hand, is triggered by cytokines like interleukin-4 (IL-4) and IL-13, resulting in an anti-inflammatory response, tissue repair, and the promotion of wound healing.

It's important to note that macrophage activation plays a crucial role in various physiological and pathological processes, including immune defense, inflammation, tissue remodeling, and even cancer progression. Dysregulation of macrophage activation has been implicated in several diseases, such as autoimmune disorders, chronic infections, and cancer.

Peritoneal macrophages are a type of immune cell that are present in the peritoneal cavity, which is the space within the abdomen that contains the liver, spleen, stomach, and intestines. These macrophages play a crucial role in the body's defense against infection and injury by engulfing and destroying foreign substances such as bacteria, viruses, and other microorganisms.

Macrophages are large phagocytic cells that originate from monocytes, which are a type of white blood cell produced in the bone marrow. When monocytes enter tissue, they can differentiate into macrophages, which have a variety of functions depending on their location and activation state.

Peritoneal macrophages are involved in various physiological processes, including the regulation of inflammation, tissue repair, and the breakdown of foreign substances. They also play a role in the development and progression of certain diseases, such as cancer and autoimmune disorders.

These macrophages can be collected from animals or humans for research purposes by injecting a solution into the peritoneal cavity and then withdrawing the fluid, which contains the macrophages. These cells can then be studied in vitro to better understand their functions and potential therapeutic targets.

Alveolar macrophages are a type of macrophage (a large phagocytic cell) that are found in the alveoli of the lungs. They play a crucial role in the immune defense system of the lungs by engulfing and destroying any foreign particles, such as dust, microorganisms, and pathogens, that enter the lungs through the process of inhalation. Alveolar macrophages also produce cytokines, which are signaling molecules that help to coordinate the immune response. They are important for maintaining the health and function of the lungs by removing debris and preventing infection.

Phagocytosis is the process by which certain cells in the body, known as phagocytes, engulf and destroy foreign particles, bacteria, or dead cells. This mechanism plays a crucial role in the immune system's response to infection and inflammation. Phagocytes, such as neutrophils, monocytes, and macrophages, have receptors on their surface that recognize and bind to specific molecules (known as antigens) on the target particles or microorganisms.

Once attached, the phagocyte extends pseudopodia (cell extensions) around the particle, forming a vesicle called a phagosome that completely encloses it. The phagosome then fuses with a lysosome, an intracellular organelle containing digestive enzymes and other chemicals. This fusion results in the formation of a phagolysosome, where the engulfed particle is broken down by the action of these enzymes, neutralizing its harmful effects and allowing for the removal of cellular debris or pathogens.

Phagocytosis not only serves as a crucial defense mechanism against infections but also contributes to tissue homeostasis by removing dead cells and debris.

Macrophage Colony-Stimulating Factor (M-CSF) is a growth factor that belongs to the family of colony-stimulating factors (CSFs). It is a glycoprotein hormone that plays a crucial role in the survival, proliferation, and differentiation of mononuclear phagocytes, including macrophages. M-CSF binds to its receptor, CSF1R, which is expressed on the surface of monocytes, macrophages, and their precursors.

M-CSF stimulates the production of mature macrophages from monocyte precursors in the bone marrow and enhances the survival and function of mature macrophages in peripheral tissues. It also promotes the activation of macrophages, increasing their ability to phagocytize and destroy foreign particles, microorganisms, and tumor cells.

In addition to its role in the immune system, M-CSF has been implicated in various physiological processes, including hematopoiesis, bone remodeling, angiogenesis, and female reproduction. Dysregulation of M-CSF signaling has been associated with several pathological conditions, such as inflammatory diseases, autoimmune disorders, and cancer.

"Cells, cultured" is a medical term that refers to cells that have been removed from an organism and grown in controlled laboratory conditions outside of the body. This process is called cell culture and it allows scientists to study cells in a more controlled and accessible environment than they would have inside the body. Cultured cells can be derived from a variety of sources, including tissues, organs, or fluids from humans, animals, or cell lines that have been previously established in the laboratory.

Cell culture involves several steps, including isolation of the cells from the tissue, purification and characterization of the cells, and maintenance of the cells in appropriate growth conditions. The cells are typically grown in specialized media that contain nutrients, growth factors, and other components necessary for their survival and proliferation. Cultured cells can be used for a variety of purposes, including basic research, drug development and testing, and production of biological products such as vaccines and gene therapies.

It is important to note that cultured cells may behave differently than they do in the body, and results obtained from cell culture studies may not always translate directly to human physiology or disease. Therefore, it is essential to validate findings from cell culture experiments using additional models and ultimately in clinical trials involving human subjects.

Lipopolysaccharides (LPS) are large molecules found in the outer membrane of Gram-negative bacteria. They consist of a hydrophilic polysaccharide called the O-antigen, a core oligosaccharide, and a lipid portion known as Lipid A. The Lipid A component is responsible for the endotoxic activity of LPS, which can trigger a powerful immune response in animals, including humans. This response can lead to symptoms such as fever, inflammation, and septic shock, especially when large amounts of LPS are introduced into the bloodstream.

C57BL/6 (C57 Black 6) is an inbred strain of laboratory mouse that is widely used in biomedical research. The term "inbred" refers to a strain of animals where matings have been carried out between siblings or other closely related individuals for many generations, resulting in a population that is highly homozygous at most genetic loci.

The C57BL/6 strain was established in 1920 by crossing a female mouse from the dilute brown (DBA) strain with a male mouse from the black strain. The resulting offspring were then interbred for many generations to create the inbred C57BL/6 strain.

C57BL/6 mice are known for their robust health, longevity, and ease of handling, making them a popular choice for researchers. They have been used in a wide range of biomedical research areas, including studies of cancer, immunology, neuroscience, cardiovascular disease, and metabolism.

One of the most notable features of the C57BL/6 strain is its sensitivity to certain genetic modifications, such as the introduction of mutations that lead to obesity or impaired glucose tolerance. This has made it a valuable tool for studying the genetic basis of complex diseases and traits.

Overall, the C57BL/6 inbred mouse strain is an important model organism in biomedical research, providing a valuable resource for understanding the genetic and molecular mechanisms underlying human health and disease.

Monocytes are a type of white blood cell that are part of the immune system. They are large cells with a round or oval shape and a nucleus that is typically indented or horseshoe-shaped. Monocytes are produced in the bone marrow and then circulate in the bloodstream, where they can differentiate into other types of immune cells such as macrophages and dendritic cells.

Monocytes play an important role in the body's defense against infection and tissue damage. They are able to engulf and digest foreign particles, microorganisms, and dead or damaged cells, which helps to clear them from the body. Monocytes also produce cytokines, which are signaling molecules that help to coordinate the immune response.

Elevated levels of monocytes in the bloodstream can be a sign of an ongoing infection, inflammation, or other medical conditions such as cancer or autoimmune disorders.

Macrophage migration-inhibitory factors (MIFs) are a group of proteins that were initially identified for their ability to inhibit the random migration of macrophages. However, subsequent research has revealed that MIFs have diverse functions in the immune system and other biological processes. They play crucial roles in inflammation, immunoregulation, and stress responses.

MIF is constitutively expressed and secreted by various cell types, including T-cells, macrophages, epithelial cells, endothelial cells, and neurons. It functions as a proinflammatory cytokine that can counteract the anti-inflammatory effects of glucocorticoids. MIF is involved in several signaling pathways and contributes to various physiological and pathophysiological processes, such as cell growth, differentiation, and survival.

Dysregulation of MIF has been implicated in numerous diseases, including autoimmune disorders, cancer, cardiovascular diseases, and neurodegenerative conditions. Therefore, understanding the functions and regulation of MIFs is essential for developing novel therapeutic strategies to target these diseases.

A cell line is a culture of cells that are grown in a laboratory for use in research. These cells are usually taken from a single cell or group of cells, and they are able to divide and grow continuously in the lab. Cell lines can come from many different sources, including animals, plants, and humans. They are often used in scientific research to study cellular processes, disease mechanisms, and to test new drugs or treatments. Some common types of human cell lines include HeLa cells (which come from a cancer patient named Henrietta Lacks), HEK293 cells (which come from embryonic kidney cells), and HUVEC cells (which come from umbilical vein endothelial cells). It is important to note that cell lines are not the same as primary cells, which are cells that are taken directly from a living organism and have not been grown in the lab.

Tumor Necrosis Factor-alpha (TNF-α) is a cytokine, a type of small signaling protein involved in immune response and inflammation. It is primarily produced by activated macrophages, although other cell types such as T-cells, natural killer cells, and mast cells can also produce it.

TNF-α plays a crucial role in the body's defense against infection and tissue injury by mediating inflammatory responses, activating immune cells, and inducing apoptosis (programmed cell death) in certain types of cells. It does this by binding to its receptors, TNFR1 and TNFR2, which are found on the surface of many cell types.

In addition to its role in the immune response, TNF-α has been implicated in the pathogenesis of several diseases, including autoimmune disorders such as rheumatoid arthritis, inflammatory bowel disease, and psoriasis, as well as cancer, where it can promote tumor growth and metastasis.

Therapeutic agents that target TNF-α, such as infliximab, adalimumab, and etanercept, have been developed to treat these conditions. However, these drugs can also increase the risk of infections and other side effects, so their use must be carefully monitored.

BALB/c is an inbred strain of laboratory mouse that is widely used in biomedical research. The strain was developed at the Institute of Cancer Research in London by Henry Baldwin and his colleagues in the 1920s, and it has since become one of the most commonly used inbred strains in the world.

BALB/c mice are characterized by their black coat color, which is determined by a recessive allele at the tyrosinase locus. They are also known for their docile and friendly temperament, making them easy to handle and work with in the laboratory.

One of the key features of BALB/c mice that makes them useful for research is their susceptibility to certain types of tumors and immune responses. For example, they are highly susceptible to developing mammary tumors, which can be induced by chemical carcinogens or viral infection. They also have a strong Th2-biased immune response, which makes them useful models for studying allergic diseases and asthma.

BALB/c mice are also commonly used in studies of genetics, neuroscience, behavior, and infectious diseases. Because they are an inbred strain, they have a uniform genetic background, which makes it easier to control for genetic factors in experiments. Additionally, because they have been bred in the laboratory for many generations, they are highly standardized and reproducible, making them ideal subjects for scientific research.

Cytokines are a broad and diverse category of small signaling proteins that are secreted by various cells, including immune cells, in response to different stimuli. They play crucial roles in regulating the immune response, inflammation, hematopoiesis, and cellular communication.

Cytokines mediate their effects by binding to specific receptors on the surface of target cells, which triggers intracellular signaling pathways that ultimately result in changes in gene expression, cell behavior, and function. Some key functions of cytokines include:

1. Regulating the activation, differentiation, and proliferation of immune cells such as T cells, B cells, natural killer (NK) cells, and macrophages.
2. Coordinating the inflammatory response by recruiting immune cells to sites of infection or tissue damage and modulating their effector functions.
3. Regulating hematopoiesis, the process of blood cell formation in the bone marrow, by controlling the proliferation, differentiation, and survival of hematopoietic stem and progenitor cells.
4. Modulating the development and function of the nervous system, including neuroinflammation, neuroprotection, and neuroregeneration.

Cytokines can be classified into several categories based on their structure, function, or cellular origin. Some common types of cytokines include interleukins (ILs), interferons (IFNs), tumor necrosis factors (TNFs), chemokines, colony-stimulating factors (CSFs), and transforming growth factors (TGFs). Dysregulation of cytokine production and signaling has been implicated in various pathological conditions, such as autoimmune diseases, chronic inflammation, cancer, and neurodegenerative disorders.

A "knockout" mouse is a genetically engineered mouse in which one or more genes have been deleted or "knocked out" using molecular biology techniques. This allows researchers to study the function of specific genes and their role in various biological processes, as well as potential associations with human diseases. The mice are generated by introducing targeted DNA modifications into embryonic stem cells, which are then used to create a live animal. Knockout mice have been widely used in biomedical research to investigate gene function, disease mechanisms, and potential therapeutic targets.

The peritoneal cavity is the potential space within the abdominal and pelvic regions, bounded by the parietal peritoneum lining the inner aspect of the abdominal and pelvic walls, and the visceral peritoneum covering the abdominal and pelvic organs. It contains a small amount of serous fluid that allows for the gliding of organs against each other during normal physiological activities such as digestion and movement. This cavity can become pathologically involved in various conditions, including inflammation, infection, hemorrhage, or neoplasia, leading to symptoms like abdominal pain, distention, or tenderness.

Thioglycolates are a group of chemical compounds that contain a thiol (sulfhydryl) group (-SH) bonded to a glycolate group. In the context of medical and cosmetic use, the term "thioglycolates" often refers to salts of thioglycolic acid, which are used as depilatories or hair-curling agents.

Thioglycolates work by breaking the disulfide bonds in keratin, the protein that makes up hair and nails. When applied to hair, thioglycolates reduce the disulfide bonds into sulfhydryl groups, making the hair more flexible and easier to shape or remove. This property is exploited in hair-curling products and depilatories (hair removal creams).

It's important to note that thioglycolates can cause skin irritation, allergic reactions, and respiratory issues in some individuals. Therefore, they should be used with caution, following the manufacturer's instructions, and in a well-ventilated area.

Macrophage Inflammatory Proteins (MIPs) are a group of chemokines, which are a type of signaling protein involved in immune responses and inflammation. Specifically, MIPs are chemotactic cytokines that attract monocytes, macrophages, and other immune cells to sites of infection or tissue damage. They play a crucial role in the recruitment and activation of these cells during the immune response.

There are several subtypes of MIPs, including MIP-1α, MIP-1β, and MIP-3α (also known as CCL3, CCL4, and CCL20, respectively). These proteins bind to specific G protein-coupled receptors on the surface of target cells, triggering a cascade of intracellular signaling events that lead to cell migration and activation.

MIPs have been implicated in a variety of inflammatory and immune-related conditions, including autoimmune diseases, cancer, and infectious diseases. They are also being studied as potential targets for the development of new therapies aimed at modulating the immune response in these conditions.

Inflammation is a complex biological response of tissues to harmful stimuli, such as pathogens, damaged cells, or irritants. It is characterized by the following signs: rubor (redness), tumor (swelling), calor (heat), dolor (pain), and functio laesa (loss of function). The process involves the activation of the immune system, recruitment of white blood cells, and release of inflammatory mediators, which contribute to the elimination of the injurious stimuli and initiation of the healing process. However, uncontrolled or chronic inflammation can also lead to tissue damage and diseases.

Antigens are substances (usually proteins) on the surface of cells, or viruses, bacteria, and other microorganisms, that can stimulate an immune response.

Differentiation in the context of myelomonocytic cells refers to the process by which these cells mature and develop into specific types of immune cells, such as monocytes, macrophages, and neutrophils.

Myelomonocytic cells are a type of white blood cell that originate from stem cells in the bone marrow. They give rise to two main types of immune cells: monocytes and granulocytes (which include neutrophils, eosinophils, and basophils).

Therefore, 'Antigens, Differentiation, Myelomonocytic' refers to the study or examination of how antigens affect the differentiation process of myelomonocytic cells into specific types of immune cells. This is an important area of research in immunology and hematology as it relates to understanding how the body responds to infections, inflammation, and cancer.

Ascitic fluid is defined as the abnormal accumulation of fluid in the peritoneal cavity, which is the space between the two layers of the peritoneum, a serous membrane that lines the abdominal cavity and covers the abdominal organs. This buildup of fluid, also known as ascites, can be caused by various medical conditions such as liver cirrhosis, cancer, heart failure, or infection. The fluid itself is typically straw-colored and clear, but it may also contain cells, proteins, and other substances depending on the underlying cause. Analysis of ascitic fluid can help doctors diagnose and manage the underlying condition causing the accumulation of fluid.

The term "Receptor, Macrophage Colony-Stimulating Factor" refers to a specific type of receptor found on the surface of certain cells, particularly macrophages and other cells involved in the immune response. This receptor binds to a protein called Macrophage Colony-Stimulating Factor (M-CSF), which is a growth factor that plays an important role in the proliferation, differentiation, and survival of mononuclear phagocytes, including macrophages.

Macrophages are key players in the immune system, responsible for engulfing and destroying foreign particles, microbes, and tumor cells. M-CSF receptor (also known as CSF1R or CD115) binds to M-CSF and activates a series of intracellular signaling pathways that promote the survival, proliferation, and differentiation of macrophages and their precursors.

Abnormalities in the M-CSF/M-CSF receptor signaling pathway have been implicated in various diseases, including cancer, inflammatory disorders, and autoimmune diseases. Therefore, targeting this pathway has emerged as a potential therapeutic strategy for these conditions.

Messenger RNA (mRNA) is a type of RNA (ribonucleic acid) that carries genetic information copied from DNA in the form of a series of three-base code "words," each of which specifies a particular amino acid. This information is used by the cell's machinery to construct proteins, a process known as translation. After being transcribed from DNA, mRNA travels out of the nucleus to the ribosomes in the cytoplasm where protein synthesis occurs. Once the protein has been synthesized, the mRNA may be degraded and recycled. Post-transcriptional modifications can also occur to mRNA, such as alternative splicing and addition of a 5' cap and a poly(A) tail, which can affect its stability, localization, and translation efficiency.

Interferon-gamma (IFN-γ) is a soluble cytokine that is primarily produced by the activation of natural killer (NK) cells and T lymphocytes, especially CD4+ Th1 cells and CD8+ cytotoxic T cells. It plays a crucial role in the regulation of the immune response against viral and intracellular bacterial infections, as well as tumor cells. IFN-γ has several functions, including activating macrophages to enhance their microbicidal activity, increasing the presentation of major histocompatibility complex (MHC) class I and II molecules on antigen-presenting cells, stimulating the proliferation and differentiation of T cells and NK cells, and inducing the production of other cytokines and chemokines. Additionally, IFN-γ has direct antiproliferative effects on certain types of tumor cells and can enhance the cytotoxic activity of immune cells against infected or malignant cells.

Clodronic acid is a medication that belongs to a class of drugs called bisphosphonates. It is used to treat and prevent osteoporosis in postmenopausal women and men with a high risk of fracture, as well as to treat Paget's disease of bone.

Clodronic acid works by inhibiting the activity of bone-resorbing cells called osteoclasts, which helps to slow down bone loss and increase bone density. This can help reduce the risk of fractures in people with osteoporosis.

The medication is available in several forms, including tablets and intravenous solutions. It is usually taken or administered once a day or once a week, depending on the specific formulation and the individual patient's needs.

Like all medications, clodronic acid can have side effects, including gastrointestinal symptoms such as nausea, vomiting, and diarrhea, as well as muscle pain, joint pain, and headaches. In rare cases, it can also cause more serious side effects such as esophageal ulcers and bone necrosis of the jaw. It is important for patients to follow their doctor's instructions carefully when taking this medication and to report any unusual symptoms or side effects promptly.

'C3H' is the name of an inbred strain of laboratory mice that was developed at the Jackson Laboratory in Bar Harbor, Maine. The mice are characterized by their uniform genetic background and have been widely used in biomedical research for many decades.

The C3H strain is particularly notable for its susceptibility to certain types of cancer, including mammary tumors and lymphomas. It also has a high incidence of age-related macular degeneration and other eye diseases. The strain is often used in studies of immunology, genetics, and carcinogenesis.

Like all inbred strains, the C3H mice are the result of many generations of brother-sister matings, which leads to a high degree of genetic uniformity within the strain. This makes them useful for studying the effects of specific genes or environmental factors on disease susceptibility and other traits. However, it also means that they may not always be representative of the genetic diversity found in outbred populations, including humans.

Scavenger receptors, class A, are a group of membrane-bound proteins found on the surface of various cell types, including macrophages, dendritic cells, and endothelial cells. These receptors play an essential role in recognizing and removing modified or damaged self and foreign molecules from the body.

Class A scavenger receptors include three members: SR-A1 (also known as Macrophage Scavenger Receptor 1 or MSR1), SR-A2 (also known as SCARA2 or MSR2), and SR-A3 (also known as SCARA3). These receptors have a wide range of ligands, including oxidized low-density lipoprotein (oxLDL), polyanionic molecules, advanced glycation end products (AGEs), and pathogens.

SR-A1 is the best characterized among the three members and has been implicated in various physiological and pathological processes, such as atherosclerosis, immune response, and neurodegenerative disorders. SR-A2 and SR-A3 have overlapping functions with SR-A1 but are less well studied.

Overall, scavenger receptors, class A, contribute to the maintenance of tissue homeostasis by clearing cellular debris and modulating immune responses. However, dysregulation of these receptors has been associated with several diseases, making them potential therapeutic targets for various pathological conditions.

A phagosome is a type of membrane-bound organelle that forms around a particle or microorganism following its engulfment by a cell, through the process of phagocytosis. This results in the formation of a vesicle containing the ingested material, which then fuses with another organelle called a lysosome to form a phago-lysosome. The lysosome contains enzymes that digest and break down the contents of the phagosome, allowing the cell to neutralize and dispose of potentially harmful substances or pathogens.

In summary, phagosomes are important organelles involved in the immune response, helping to protect the body against infection and disease.

'Gene expression regulation' refers to the processes that control whether, when, and where a particular gene is expressed, meaning the production of a specific protein or functional RNA encoded by that gene. This complex mechanism can be influenced by various factors such as transcription factors, chromatin remodeling, DNA methylation, non-coding RNAs, and post-transcriptional modifications, among others. Proper regulation of gene expression is crucial for normal cellular function, development, and maintaining homeostasis in living organisms. Dysregulation of gene expression can lead to various diseases, including cancer and genetic disorders.

Nitric oxide (NO) is a molecule made up of one nitrogen atom and one oxygen atom. In the body, it is a crucial signaling molecule involved in various physiological processes such as vasodilation, immune response, neurotransmission, and inhibition of platelet aggregation. It is produced naturally by the enzyme nitric oxide synthase (NOS) from the amino acid L-arginine. Inhaled nitric oxide is used medically to treat pulmonary hypertension in newborns and adults, as it helps to relax and widen blood vessels, improving oxygenation and blood flow.

Pulmonary alveoli, also known as air sacs, are tiny clusters of air-filled pouches located at the end of the bronchioles in the lungs. They play a crucial role in the process of gas exchange during respiration. The thin walls of the alveoli, called alveolar membranes, allow oxygen from inhaled air to pass into the bloodstream and carbon dioxide from the bloodstream to pass into the alveoli to be exhaled out of the body. This vital function enables the lungs to supply oxygen-rich blood to the rest of the body and remove waste products like carbon dioxide.

Nitric Oxide Synthase Type II (NOS2), also known as Inducible Nitric Oxide Synthase (iNOS), is an enzyme that catalyzes the production of nitric oxide (NO) from L-arginine. Unlike other isoforms of NOS, NOS2 is not constitutively expressed and its expression can be induced by various stimuli such as cytokines, lipopolysaccharides, and bacterial products. Once induced, NOS2 produces large amounts of NO, which plays a crucial role in the immune response against invading pathogens. However, excessive or prolonged production of NO by NOS2 has been implicated in various pathological conditions such as inflammation, septic shock, and neurodegenerative disorders.

Signal transduction is the process by which a cell converts an extracellular signal, such as a hormone or neurotransmitter, into an intracellular response. This involves a series of molecular events that transmit the signal from the cell surface to the interior of the cell, ultimately resulting in changes in gene expression, protein activity, or metabolism.

The process typically begins with the binding of the extracellular signal to a receptor located on the cell membrane. This binding event activates the receptor, which then triggers a cascade of intracellular signaling molecules, such as second messengers, protein kinases, and ion channels. These molecules amplify and propagate the signal, ultimately leading to the activation or inhibition of specific cellular responses.

Signal transduction pathways are highly regulated and can be modulated by various factors, including other signaling molecules, post-translational modifications, and feedback mechanisms. Dysregulation of these pathways has been implicated in a variety of diseases, including cancer, diabetes, and neurological disorders.

The spleen is an organ in the upper left side of the abdomen, next to the stomach and behind the ribs. It plays multiple supporting roles in the body:

1. It fights infection by acting as a filter for the blood. Old red blood cells are recycled in the spleen, and platelets and white blood cells are stored there.
2. The spleen also helps to control the amount of blood in the body by removing excess red blood cells and storing platelets.
3. It has an important role in immune function, producing antibodies and removing microorganisms and damaged red blood cells from the bloodstream.

The spleen can be removed without causing any significant problems, as other organs take over its functions. This is known as a splenectomy and may be necessary if the spleen is damaged or diseased.

Macrophage-activating factors (MAFs) are substances that stimulate the activation and function of macrophages, which are a type of white blood cell involved in the immune response. These factors can be produced by various cells, including T lymphocytes, and can enhance the ability of macrophages to phagocytize (ingest and destroy) foreign substances, such as bacteria and viruses, and to produce cytokines, which are signaling molecules that mediate and regulate the immune response.

MAFs can be classified into two main groups: endogenous and exogenous. Endogenous MAFs are produced by cells of the body in response to various stimuli, such as infection or inflammation. Examples of endogenous MAFs include interferon-gamma (IFN-γ), granulocyte-macrophage colony-stimulating factor (GM-CSF), and tumor necrosis factor-alpha (TNF-α). Exogenous MAFs, on the other hand, are substances that are introduced into the body from outside sources, such as bacterial toxins or synthetic compounds, and can also activate macrophages.

MAFs play an important role in the immune response by helping to coordinate the activities of different types of immune cells and regulate the intensity and duration of the immune response. Dysregulation of MAF production or activity has been implicated in various diseases, including autoimmune disorders, chronic infections, and cancer.

Zymosan is a type of substance that is derived from the cell walls of yeast and some types of fungi. It's often used in laboratory research as an agent to stimulate inflammation, because it can activate certain immune cells (such as neutrophils) and cause them to release pro-inflammatory chemicals.

In medical terms, Zymosan is sometimes used as a tool for studying the immune system and inflammation in experimental settings. It's important to note that Zymosan itself is not a medical condition or disease, but rather a research reagent with potential applications in understanding human health and disease.

Bone marrow cells are the types of cells found within the bone marrow, which is the spongy tissue inside certain bones in the body. The main function of bone marrow is to produce blood cells. There are two types of bone marrow: red and yellow. Red bone marrow is where most blood cell production takes place, while yellow bone marrow serves as a fat storage site.

The three main types of bone marrow cells are:

1. Hematopoietic stem cells (HSCs): These are immature cells that can differentiate into any type of blood cell, including red blood cells, white blood cells, and platelets. They have the ability to self-renew, meaning they can divide and create more hematopoietic stem cells.
2. Red blood cell progenitors: These are immature cells that will develop into mature red blood cells, also known as erythrocytes. Red blood cells carry oxygen from the lungs to the body's tissues and carbon dioxide back to the lungs.
3. Myeloid and lymphoid white blood cell progenitors: These are immature cells that will develop into various types of white blood cells, which play a crucial role in the body's immune system by fighting infections and diseases. Myeloid progenitors give rise to granulocytes (neutrophils, eosinophils, and basophils), monocytes, and megakaryocytes (which eventually become platelets). Lymphoid progenitors differentiate into B cells, T cells, and natural killer (NK) cells.

Bone marrow cells are essential for maintaining a healthy blood cell count and immune system function. Abnormalities in bone marrow cells can lead to various medical conditions, such as anemia, leukopenia, leukocytosis, thrombocytopenia, or thrombocytosis, depending on the specific type of blood cell affected. Additionally, bone marrow cells are often used in transplantation procedures to treat patients with certain types of cancer, such as leukemia and lymphoma, or other hematologic disorders.

Low-density lipoproteins (LDL), also known as "bad cholesterol," are a type of lipoprotein that carry cholesterol and other fats from the liver to cells throughout the body. High levels of LDL in the blood can lead to the buildup of cholesterol in the walls of the arteries, which can increase the risk of heart disease and stroke.

Lipoproteins are complex particles composed of proteins (apolipoproteins) and lipids (cholesterol, triglycerides, and phospholipids) that are responsible for transporting fat molecules around the body in the bloodstream. LDL is one type of lipoprotein, along with high-density lipoproteins (HDL), very low-density lipoproteins (VLDL), and chylomicrons.

LDL particles are smaller than HDL particles and can easily penetrate the artery walls, leading to the formation of plaques that can narrow or block the arteries. Therefore, maintaining healthy levels of LDL in the blood is essential for preventing cardiovascular disease.

Mannose-binding lectins (MBLs) are a group of proteins that belong to the collectin family and play a crucial role in the innate immune system. They are primarily produced by the liver and secreted into the bloodstream. MBLs have a specific affinity for mannose sugar residues found on the surface of various microorganisms, including bacteria, viruses, fungi, and parasites.

The primary function of MBLs is to recognize and bind to these mannose-rich structures, which triggers the complement system's activation through the lectin pathway. This process leads to the destruction of the microorganism by opsonization (coating the microbe to enhance phagocytosis) or direct lysis. MBLs also have the ability to neutralize certain viruses and inhibit the replication of others, further contributing to their antimicrobial activity.

Deficiencies in MBL levels or function have been associated with an increased susceptibility to infections, particularly in children and older adults. However, the clinical significance of MBL deficiency remains a subject of ongoing research.

Chemokine (C-C motif) ligand 2, also known as monocyte chemoattractant protein-1 (MCP-1), is a small signaling protein that belongs to the chemokine family. Chemokines are a group of cytokines, or regulatory proteins, that play important roles in immune responses and inflammation by recruiting various immune cells to sites of infection or injury.

CCL2 specifically acts as a chemoattractant for monocytes, memory T cells, and dendritic cells, guiding them to migrate towards the source of infection or tissue damage. It does this by binding to its receptor, CCR2, which is expressed on the surface of these immune cells.

CCL2 has been implicated in several pathological conditions, including atherosclerosis, rheumatoid arthritis, and various cancers, where it contributes to the recruitment of immune cells that can exacerbate tissue damage or promote tumor growth and metastasis. Therefore, targeting CCL2 or its signaling pathways has emerged as a potential therapeutic strategy for these diseases.

Cell differentiation is the process by which a less specialized cell, or stem cell, becomes a more specialized cell type with specific functions and structures. This process involves changes in gene expression, which are regulated by various intracellular signaling pathways and transcription factors. Differentiation results in the development of distinct cell types that make up tissues and organs in multicellular organisms. It is a crucial aspect of embryonic development, tissue repair, and maintenance of homeostasis in the body.

A lung is a pair of spongy, elastic organs in the chest that work together to enable breathing. They are responsible for taking in oxygen and expelling carbon dioxide through the process of respiration. The left lung has two lobes, while the right lung has three lobes. The lungs are protected by the ribcage and are covered by a double-layered membrane called the pleura. The trachea divides into two bronchi, which further divide into smaller bronchioles, leading to millions of tiny air sacs called alveoli, where the exchange of gases occurs.

Scavenger receptors are a class of cell surface receptors that play a crucial role in the recognition and clearance of various biomolecules, including modified self-molecules, pathogens, and apoptotic cells. These receptors are expressed mainly by phagocytic cells such as macrophages and dendritic cells, but they can also be found on other cell types, including endothelial cells and smooth muscle cells.

Scavenger receptors have broad specificity and can bind to a wide range of ligands, including oxidized low-density lipoprotein (oxLDL), polyanionic molecules, advanced glycation end products (AGEs), and pathogen-associated molecular patterns (PAMPs). The binding of ligands to scavenger receptors triggers various cellular responses, such as phagocytosis, endocytosis, signaling cascades, and the production of cytokines and chemokines.

Scavenger receptors are classified into several families based on their structural features and ligand specificity, including:

1. Class A (SR-A): This family includes SR-AI, SR-AII, and MARCO, which bind to oxLDL, bacteria, and apoptotic cells.
2. Class B (SR-B): This family includes SR-BI, CD36, and LIMPII, which bind to lipoproteins, phospholipids, and pathogens.
3. Class C (SR-C): This family includes DEC-205, MRC1, and LOX-1, which bind to various ligands, including apoptotic cells, bacteria, and oxLDL.
4. Class D (SR-D): This family includes SCARF1, which binds to PAMPs and damage-associated molecular patterns (DAMPs).
5. Class E (SR-E): This family includes CXCL16, which binds to chemokine CXCR6 and phosphatidylserine.

Scavenger receptors play a critical role in maintaining tissue homeostasis by removing damaged or altered molecules and cells, modulating immune responses, and regulating lipid metabolism. Dysregulation of scavenger receptor function has been implicated in various pathological conditions, including atherosclerosis, inflammation, infection, and cancer.

Flow cytometry is a medical and research technique used to measure physical and chemical characteristics of cells or particles, one cell at a time, as they flow in a fluid stream through a beam of light. The properties measured include:

* Cell size (light scatter)
* Cell internal complexity (granularity, also light scatter)
* Presence or absence of specific proteins or other molecules on the cell surface or inside the cell (using fluorescent antibodies or other fluorescent probes)

The technique is widely used in cell counting, cell sorting, protein engineering, biomarker discovery and monitoring disease progression, particularly in hematology, immunology, and cancer research.

NF-κB (Nuclear Factor kappa-light-chain-enhancer of activated B cells) is a protein complex that plays a crucial role in regulating the immune response to infection and inflammation, as well as in cell survival, differentiation, and proliferation. It is composed of several subunits, including p50, p52, p65 (RelA), c-Rel, and RelB, which can form homodimers or heterodimers that bind to specific DNA sequences called κB sites in the promoter regions of target genes.

Under normal conditions, NF-κB is sequestered in the cytoplasm by inhibitory proteins known as IκBs (inhibitors of κB). However, upon stimulation by various signals such as cytokines, bacterial or viral products, and stress, IκBs are phosphorylated, ubiquitinated, and degraded, leading to the release and activation of NF-κB. Activated NF-κB then translocates to the nucleus, where it binds to κB sites and regulates the expression of target genes involved in inflammation, immunity, cell survival, and proliferation.

Dysregulation of NF-κB signaling has been implicated in various pathological conditions such as cancer, chronic inflammation, autoimmune diseases, and neurodegenerative disorders. Therefore, targeting NF-κB signaling has emerged as a potential therapeutic strategy for the treatment of these diseases.

Toll-Like Receptor 4 (TLR4) is a type of protein found on the surface of some cells in the human body, including immune cells like macrophages and dendritic cells. It belongs to a class of proteins called pattern recognition receptors (PRRs), which play a crucial role in the innate immune system's response to infection.

TLR4 recognizes and responds to specific molecules found on gram-negative bacteria, such as lipopolysaccharide (LPS), also known as endotoxin. When TLR4 binds to LPS, it triggers a signaling cascade that leads to the activation of immune cells, production of pro-inflammatory cytokines and chemokines, and initiation of the adaptive immune response.

TLR4 is an essential component of the body's defense against gram-negative bacterial infections, but its overactivation can also contribute to the development of various inflammatory diseases, such as sepsis, atherosclerosis, and certain types of cancer.

Cell surface receptors, also known as membrane receptors, are proteins located on the cell membrane that bind to specific molecules outside the cell, known as ligands. These receptors play a crucial role in signal transduction, which is the process of converting an extracellular signal into an intracellular response.

Cell surface receptors can be classified into several categories based on their structure and mechanism of action, including:

1. Ion channel receptors: These receptors contain a pore that opens to allow ions to flow across the cell membrane when they bind to their ligands. This ion flux can directly activate or inhibit various cellular processes.
2. G protein-coupled receptors (GPCRs): These receptors consist of seven transmembrane domains and are associated with heterotrimeric G proteins that modulate intracellular signaling pathways upon ligand binding.
3. Enzyme-linked receptors: These receptors possess an intrinsic enzymatic activity or are linked to an enzyme, which becomes activated when the receptor binds to its ligand. This activation can lead to the initiation of various signaling cascades within the cell.
4. Receptor tyrosine kinases (RTKs): These receptors contain intracellular tyrosine kinase domains that become activated upon ligand binding, leading to the phosphorylation and activation of downstream signaling molecules.
5. Integrins: These receptors are transmembrane proteins that mediate cell-cell or cell-matrix interactions by binding to extracellular matrix proteins or counter-receptors on adjacent cells. They play essential roles in cell adhesion, migration, and survival.

Cell surface receptors are involved in various physiological processes, including neurotransmission, hormone signaling, immune response, and cell growth and differentiation. Dysregulation of these receptors can contribute to the development of numerous diseases, such as cancer, diabetes, and neurological disorders.

Atherosclerosis is a medical condition characterized by the buildup of plaques, made up of fat, cholesterol, calcium, and other substances found in the blood, on the inner walls of the arteries. This process gradually narrows and hardens the arteries, reducing the flow of oxygen-rich blood to various parts of the body. Atherosclerosis can affect any artery in the body, including those that supply blood to the heart (coronary arteries), brain, limbs, and other organs. The progressive narrowing and hardening of the arteries can lead to serious complications such as coronary artery disease, carotid artery disease, peripheral artery disease, and aneurysms, which can result in heart attacks, strokes, or even death if left untreated.

The exact cause of atherosclerosis is not fully understood, but it is believed to be associated with several risk factors, including high blood pressure, high cholesterol levels, smoking, diabetes, obesity, physical inactivity, and a family history of the condition. Atherosclerosis can often progress without any symptoms for many years, but as the disease advances, it can lead to various signs and symptoms depending on which arteries are affected. Treatment typically involves lifestyle changes, medications, and, in some cases, surgical procedures to restore blood flow.

Animal disease models are specialized animals, typically rodents such as mice or rats, that have been genetically engineered or exposed to certain conditions to develop symptoms and physiological changes similar to those seen in human diseases. These models are used in medical research to study the pathophysiology of diseases, identify potential therapeutic targets, test drug efficacy and safety, and understand disease mechanisms.

The genetic modifications can include knockout or knock-in mutations, transgenic expression of specific genes, or RNA interference techniques. The animals may also be exposed to environmental factors such as chemicals, radiation, or infectious agents to induce the disease state.

Examples of animal disease models include:

1. Mouse models of cancer: Genetically engineered mice that develop various types of tumors, allowing researchers to study cancer initiation, progression, and metastasis.
2. Alzheimer's disease models: Transgenic mice expressing mutant human genes associated with Alzheimer's disease, which exhibit amyloid plaque formation and cognitive decline.
3. Diabetes models: Obese and diabetic mouse strains like the NOD (non-obese diabetic) or db/db mice, used to study the development of type 1 and type 2 diabetes, respectively.
4. Cardiovascular disease models: Atherosclerosis-prone mice, such as ApoE-deficient or LDLR-deficient mice, that develop plaque buildup in their arteries when fed a high-fat diet.
5. Inflammatory bowel disease models: Mice with genetic mutations affecting intestinal barrier function and immune response, such as IL-10 knockout or SAMP1/YitFc mice, which develop colitis.

Animal disease models are essential tools in preclinical research, but it is important to recognize their limitations. Differences between species can affect the translatability of results from animal studies to human patients. Therefore, researchers must carefully consider the choice of model and interpret findings cautiously when applying them to human diseases.

Immunohistochemistry (IHC) is a technique used in pathology and laboratory medicine to identify specific proteins or antigens in tissue sections. It combines the principles of immunology and histology to detect the presence and location of these target molecules within cells and tissues. This technique utilizes antibodies that are specific to the protein or antigen of interest, which are then tagged with a detection system such as a chromogen or fluorophore. The stained tissue sections can be examined under a microscope, allowing for the visualization and analysis of the distribution and expression patterns of the target molecule in the context of the tissue architecture. Immunohistochemistry is widely used in diagnostic pathology to help identify various diseases, including cancer, infectious diseases, and immune-mediated disorders.

Innate immunity, also known as non-specific immunity or natural immunity, is the inherent defense mechanism that provides immediate protection against potentially harmful pathogens (like bacteria, viruses, fungi, and parasites) without the need for prior exposure. This type of immunity is present from birth and does not adapt to specific threats over time.

Innate immune responses involve various mechanisms such as:

1. Physical barriers: Skin and mucous membranes prevent pathogens from entering the body.
2. Chemical barriers: Enzymes, stomach acid, and lysozyme in tears, saliva, and sweat help to destroy or inhibit the growth of microorganisms.
3. Cellular responses: Phagocytic cells (neutrophils, monocytes, macrophages) recognize and engulf foreign particles and pathogens, while natural killer (NK) cells target and eliminate virus-infected or cancerous cells.
4. Inflammatory response: When an infection occurs, the innate immune system triggers inflammation to increase blood flow, recruit immune cells, and remove damaged tissue.
5. Complement system: A group of proteins that work together to recognize and destroy pathogens directly or enhance phagocytosis by coating them with complement components (opsonization).

Innate immunity plays a crucial role in initiating the adaptive immune response, which is specific to particular pathogens and provides long-term protection through memory cells. Both innate and adaptive immunity work together to maintain overall immune homeostasis and protect the body from infections and diseases.

Neutrophils are a type of white blood cell that are part of the immune system's response to infection. They are produced in the bone marrow and released into the bloodstream where they circulate and are able to move quickly to sites of infection or inflammation in the body. Neutrophils are capable of engulfing and destroying bacteria, viruses, and other foreign substances through a process called phagocytosis. They are also involved in the release of inflammatory mediators, which can contribute to tissue damage in some cases. Neutrophils are characterized by the presence of granules in their cytoplasm, which contain enzymes and other proteins that help them carry out their immune functions.

Interleukin-6 (IL-6) is a cytokine, a type of protein that plays a crucial role in communication between cells, especially in the immune system. It is produced by various cells including T-cells, B-cells, fibroblasts, and endothelial cells in response to infection, injury, or inflammation.

IL-6 has diverse effects on different cell types. In the immune system, it stimulates the growth and differentiation of B-cells into plasma cells that produce antibodies. It also promotes the activation and survival of T-cells. Moreover, IL-6 plays a role in fever induction by acting on the hypothalamus to raise body temperature during an immune response.

In addition to its functions in the immune system, IL-6 has been implicated in various physiological processes such as hematopoiesis (the formation of blood cells), bone metabolism, and neural development. However, abnormal levels of IL-6 have also been associated with several diseases, including autoimmune disorders, chronic inflammation, and cancer.

Immunologic receptors are specialized proteins found on the surface of immune cells that recognize and bind to specific molecules, known as antigens, on the surface of pathogens or infected cells. This binding triggers a series of intracellular signaling events that activate the immune cell and initiate an immune response.

There are several types of immunologic receptors, including:

1. T-cell receptors (TCRs): These receptors are found on the surface of T cells and recognize antigens presented in the context of major histocompatibility complex (MHC) molecules.
2. B-cell receptors (BCRs): These receptors are found on the surface of B cells and recognize free antigens in solution.
3. Pattern recognition receptors (PRRs): These receptors are found inside immune cells and recognize conserved molecular patterns associated with pathogens, such as lipopolysaccharides and flagellin.
4. Fc receptors: These receptors are found on the surface of various immune cells and bind to the constant region of antibodies, mediating effector functions such as phagocytosis and antibody-dependent cellular cytotoxicity (ADCC).

Immunologic receptors play a critical role in the recognition and elimination of pathogens and infected cells, and dysregulation of these receptors can lead to immune disorders and diseases.

Interleukin-1 (IL-1) is a type of cytokine, which are proteins that play a crucial role in cell signaling. Specifically, IL-1 is a pro-inflammatory cytokine that is involved in the regulation of immune and inflammatory responses in the body. It is produced by various cells, including monocytes, macrophages, and dendritic cells, in response to infection or injury.

IL-1 exists in two forms, IL-1α and IL-1β, which have similar biological activities but are encoded by different genes. Both forms of IL-1 bind to the same receptor, IL-1R, and activate intracellular signaling pathways that lead to the production of other cytokines, chemokines, and inflammatory mediators.

IL-1 has a wide range of biological effects, including fever induction, activation of immune cells, regulation of hematopoiesis (the formation of blood cells), and modulation of bone metabolism. Dysregulation of IL-1 production or activity has been implicated in various inflammatory diseases, such as rheumatoid arthritis, gout, and inflammatory bowel disease. Therefore, IL-1 is an important target for the development of therapies aimed at modulating the immune response and reducing inflammation.

Inflammation mediators are substances that are released by the body in response to injury or infection, which contribute to the inflammatory response. These mediators include various chemical factors such as cytokines, chemokines, prostaglandins, leukotrienes, and histamine, among others. They play a crucial role in regulating the inflammatory process by attracting immune cells to the site of injury or infection, increasing blood flow to the area, and promoting the repair and healing of damaged tissues. However, an overactive or chronic inflammatory response can also contribute to the development of various diseases and conditions, such as autoimmune disorders, cardiovascular disease, and cancer.

Apoptosis is a programmed and controlled cell death process that occurs in multicellular organisms. It is a natural process that helps maintain tissue homeostasis by eliminating damaged, infected, or unwanted cells. During apoptosis, the cell undergoes a series of morphological changes, including cell shrinkage, chromatin condensation, and fragmentation into membrane-bound vesicles called apoptotic bodies. These bodies are then recognized and engulfed by neighboring cells or phagocytic cells, preventing an inflammatory response. Apoptosis is regulated by a complex network of intracellular signaling pathways that involve proteins such as caspases, Bcl-2 family members, and inhibitors of apoptosis (IAPs).

Granulocyte-Macrophage Colony-Stimulating Factor (GM-CSF) is a type of cytokine, which is a small signaling protein involved in immune response and hematopoiesis (the formation of blood cells). GM-CSF's specific role is to stimulate the production, proliferation, and activation of granulocytes (a type of white blood cell that fights against infection) and macrophages (large white blood cells that eat foreign substances, bacteria, and dead or dying cells).

In medical terms, GM-CSF is often used in therapeutic settings to boost the production of white blood cells in patients undergoing chemotherapy or radiation treatment for cancer. This can help to reduce the risk of infection during these treatments. It can also be used to promote the growth and differentiation of stem cells in bone marrow transplant procedures.

T-lymphocytes, also known as T-cells, are a type of white blood cell that plays a key role in the adaptive immune system's response to infection. They are produced in the bone marrow and mature in the thymus gland. There are several different types of T-cells, including CD4+ helper T-cells, CD8+ cytotoxic T-cells, and regulatory T-cells (Tregs).

CD4+ helper T-cells assist in activating other immune cells, such as B-lymphocytes and macrophages. They also produce cytokines, which are signaling molecules that help coordinate the immune response. CD8+ cytotoxic T-cells directly kill infected cells by releasing toxic substances. Regulatory T-cells help maintain immune tolerance and prevent autoimmune diseases by suppressing the activity of other immune cells.

T-lymphocytes are important in the immune response to viral infections, cancer, and other diseases. Dysfunction or depletion of T-cells can lead to immunodeficiency and increased susceptibility to infections. On the other hand, an overactive T-cell response can contribute to autoimmune diseases and chronic inflammation.

CD (cluster of differentiation) antigens are cell-surface proteins that are expressed on leukocytes (white blood cells) and can be used to identify and distinguish different subsets of these cells. They are important markers in the field of immunology and hematology, and are commonly used to diagnose and monitor various diseases, including cancer, autoimmune disorders, and infectious diseases.

CD antigens are designated by numbers, such as CD4, CD8, CD19, etc., which refer to specific proteins found on the surface of different types of leukocytes. For example, CD4 is a protein found on the surface of helper T cells, while CD8 is found on cytotoxic T cells.

CD antigens can be used as targets for immunotherapy, such as monoclonal antibody therapy, in which antibodies are designed to bind to specific CD antigens and trigger an immune response against cancer cells or infected cells. They can also be used as markers to monitor the effectiveness of treatments and to detect minimal residual disease (MRD) after treatment.

It's important to note that not all CD antigens are exclusive to leukocytes, some can be found on other cell types as well, and their expression can vary depending on the activation state or differentiation stage of the cells.

Interleukin-10 (IL-10) is an anti-inflammatory cytokine that plays a crucial role in the modulation of immune responses. It is produced by various cell types, including T cells, macrophages, and dendritic cells. IL-10 inhibits the production of pro-inflammatory cytokines, such as TNF-α, IL-1, IL-6, IL-8, and IL-12, and downregulates the expression of costimulatory molecules on antigen-presenting cells. This results in the suppression of T cell activation and effector functions, which ultimately helps to limit tissue damage during inflammation and promote tissue repair. Dysregulation of IL-10 has been implicated in various pathological conditions, including chronic infections, autoimmune diseases, and cancer.

In the field of medicine, "time factors" refer to the duration of symptoms or time elapsed since the onset of a medical condition, which can have significant implications for diagnosis and treatment. Understanding time factors is crucial in determining the progression of a disease, evaluating the effectiveness of treatments, and making critical decisions regarding patient care.

For example, in stroke management, "time is brain," meaning that rapid intervention within a specific time frame (usually within 4.5 hours) is essential to administering tissue plasminogen activator (tPA), a clot-busting drug that can minimize brain damage and improve patient outcomes. Similarly, in trauma care, the "golden hour" concept emphasizes the importance of providing definitive care within the first 60 minutes after injury to increase survival rates and reduce morbidity.

Time factors also play a role in monitoring the progression of chronic conditions like diabetes or heart disease, where regular follow-ups and assessments help determine appropriate treatment adjustments and prevent complications. In infectious diseases, time factors are crucial for initiating antibiotic therapy and identifying potential outbreaks to control their spread.

Overall, "time factors" encompass the significance of recognizing and acting promptly in various medical scenarios to optimize patient outcomes and provide effective care.

Inbred strains of mice are defined as lines of mice that have been brother-sister mated for at least 20 consecutive generations. This results in a high degree of homozygosity, where the mice of an inbred strain are genetically identical to one another, with the exception of spontaneous mutations.

Inbred strains of mice are widely used in biomedical research due to their genetic uniformity and stability, which makes them useful for studying the genetic basis of various traits, diseases, and biological processes. They also provide a consistent and reproducible experimental system, as compared to outbred or genetically heterogeneous populations.

Some commonly used inbred strains of mice include C57BL/6J, BALB/cByJ, DBA/2J, and 129SvEv. Each strain has its own unique genetic background and phenotypic characteristics, which can influence the results of experiments. Therefore, it is important to choose the appropriate inbred strain for a given research question.

Reverse Transcriptase Polymerase Chain Reaction (RT-PCR) is a laboratory technique used in molecular biology to amplify and detect specific DNA sequences. This technique is particularly useful for the detection and quantification of RNA viruses, as well as for the analysis of gene expression.

The process involves two main steps: reverse transcription and polymerase chain reaction (PCR). In the first step, reverse transcriptase enzyme is used to convert RNA into complementary DNA (cDNA) by reading the template provided by the RNA molecule. This cDNA then serves as a template for the PCR amplification step.

In the second step, the PCR reaction uses two primers that flank the target DNA sequence and a thermostable polymerase enzyme to repeatedly copy the targeted cDNA sequence. The reaction mixture is heated and cooled in cycles, allowing the primers to anneal to the template, and the polymerase to extend the new strand. This results in exponential amplification of the target DNA sequence, making it possible to detect even small amounts of RNA or cDNA.

RT-PCR is a sensitive and specific technique that has many applications in medical research and diagnostics, including the detection of viruses such as HIV, hepatitis C virus, and SARS-CoV-2 (the virus that causes COVID-19). It can also be used to study gene expression, identify genetic mutations, and diagnose genetic disorders.

Bronchoalveolar lavage (BAL) fluid is a type of clinical specimen obtained through a procedure called bronchoalveolar lavage. This procedure involves inserting a bronchoscope into the lungs and instilling a small amount of saline solution into a specific area of the lung, then gently aspirating the fluid back out. The fluid that is recovered is called bronchoalveolar lavage fluid.

BAL fluid contains cells and other substances that are present in the lower respiratory tract, including the alveoli (the tiny air sacs where gas exchange occurs). By analyzing BAL fluid, doctors can diagnose various lung conditions, such as pneumonia, interstitial lung disease, and lung cancer. They can also monitor the effectiveness of treatments for these conditions by comparing the composition of BAL fluid before and after treatment.

BAL fluid is typically analyzed for its cellular content, including the number and type of white blood cells present, as well as for the presence of bacteria, viruses, or other microorganisms. The fluid may also be tested for various proteins, enzymes, and other biomarkers that can provide additional information about lung health and disease.

Chemokines are a family of small cytokines, or signaling proteins, that are secreted by cells and play an important role in the immune system. They are chemotactic, meaning they can attract and guide the movement of various immune cells to specific locations within the body. Chemokines do this by binding to G protein-coupled receptors on the surface of target cells, initiating a signaling cascade that leads to cell migration.

There are four main subfamilies of chemokines, classified based on the arrangement of conserved cysteine residues near the amino terminus: CXC, CC, C, and CX3C. Different chemokines have specific roles in inflammation, immune surveillance, hematopoiesis, and development. Dysregulation of chemokine function has been implicated in various diseases, including autoimmune disorders, infections, and cancer.

In summary, Chemokines are a group of signaling proteins that play a crucial role in the immune system by directing the movement of immune cells to specific locations within the body, thus helping to coordinate the immune response.

Lysosomes are membrane-bound organelles found in the cytoplasm of eukaryotic cells. They are responsible for breaking down and recycling various materials, such as waste products, foreign substances, and damaged cellular components, through a process called autophagy or phagocytosis. Lysosomes contain hydrolytic enzymes that can break down biomolecules like proteins, nucleic acids, lipids, and carbohydrates into their basic building blocks, which can then be reused by the cell. They play a crucial role in maintaining cellular homeostasis and are often referred to as the "garbage disposal system" of the cell.

Up-regulation is a term used in molecular biology and medicine to describe an increase in the expression or activity of a gene, protein, or receptor in response to a stimulus. This can occur through various mechanisms such as increased transcription, translation, or reduced degradation of the molecule. Up-regulation can have important functional consequences, for example, enhancing the sensitivity or response of a cell to a hormone, neurotransmitter, or drug. It is a normal physiological process that can also be induced by disease or pharmacological interventions.

Phagocytes are a type of white blood cell in the immune system that engulf and destroy foreign particles, microbes, and cellular debris. They play a crucial role in the body's defense against infection and tissue damage. There are several types of phagocytes, including neutrophils, monocytes, macrophages, and dendritic cells. These cells have receptors that recognize and bind to specific molecules on the surface of foreign particles or microbes, allowing them to engulf and digest the invaders. Phagocytosis is an important mechanism for maintaining tissue homeostasis and preventing the spread of infection.

Chemokine (C-C motif) ligand 4, also known as CCL4 or MIP-1β (Macrophage Inflammatory Protein-1β), is a small signaling protein that belongs to the chemokine family. Chemokines are a group of cytokines, or regulatory proteins, that play crucial roles in immunity and inflammation by directing the migration of various immune cells to sites of infection, injury, or tissue damage.

CCL4 is produced primarily by T cells, monocytes, macrophages, and dendritic cells. It exerts its functions by binding to specific chemokine receptors found on the surface of target cells, particularly CCR5 and CXCR3. The primary role of CCL4 is to recruit immune cells like T cells, eosinophils, and monocytes/macrophages to areas of inflammation or infection, where it contributes to the elimination of pathogens and facilitates tissue repair.

Aberrant regulation of chemokines, including CCL4, has been implicated in various disease conditions such as chronic inflammation, autoimmune disorders, and viral infections like HIV. In HIV infection, CCL4 plays a significant role in the viral replication and pathogenesis by acting as a co-receptor for virus entry into host cells.

Chemokine (C-C motif) ligand 3 (CCL3), also known as macrophage inflammatory protein-1 alpha (MIP-1α), is a small signaling protein belonging to the chemokine family. Chemokines are a group of cytokines, or cell signaling molecules, that play important roles in immune responses and inflammation. They mediate their effects by interacting with specific receptors on the surface of target cells, leading to various biological responses such as chemotaxis (directed migration) of immune cells.

CCL3 is primarily produced by activated T cells, monocytes, macrophages, and other immune cells in response to infection or injury. It plays a crucial role in recruiting immune cells like monocytes, neutrophils, and dendritic cells to the sites of inflammation or infection. CCL3 also contributes to the activation and differentiation of immune cells, thereby participating in the regulation of adaptive immunity. Dysregulation of CCL3 has been implicated in several pathological conditions, including autoimmune diseases, chronic inflammation, and cancer.

Recombinant proteins are artificially created proteins produced through the use of recombinant DNA technology. This process involves combining DNA molecules from different sources to create a new set of genes that encode for a specific protein. The resulting recombinant protein can then be expressed, purified, and used for various applications in research, medicine, and industry.

Recombinant proteins are widely used in biomedical research to study protein function, structure, and interactions. They are also used in the development of diagnostic tests, vaccines, and therapeutic drugs. For example, recombinant insulin is a common treatment for diabetes, while recombinant human growth hormone is used to treat growth disorders.

The production of recombinant proteins typically involves the use of host cells, such as bacteria, yeast, or mammalian cells, which are engineered to express the desired protein. The host cells are transformed with a plasmid vector containing the gene of interest, along with regulatory elements that control its expression. Once the host cells are cultured and the protein is expressed, it can be purified using various chromatography techniques.

Overall, recombinant proteins have revolutionized many areas of biology and medicine, enabling researchers to study and manipulate proteins in ways that were previously impossible.

Gene expression is the process by which the information encoded in a gene is used to synthesize a functional gene product, such as a protein or RNA molecule. This process involves several steps: transcription, RNA processing, and translation. During transcription, the genetic information in DNA is copied into a complementary RNA molecule, known as messenger RNA (mRNA). The mRNA then undergoes RNA processing, which includes adding a cap and tail to the mRNA and splicing out non-coding regions called introns. The resulting mature mRNA is then translated into a protein on ribosomes in the cytoplasm through the process of translation.

The regulation of gene expression is a complex and highly controlled process that allows cells to respond to changes in their environment, such as growth factors, hormones, and stress signals. This regulation can occur at various stages of gene expression, including transcriptional activation or repression, RNA processing, mRNA stability, and translation. Dysregulation of gene expression has been implicated in many diseases, including cancer, genetic disorders, and neurological conditions.

Arginase is an enzyme that plays a role in the metabolism of arginine, an amino acid. It works by breaking down arginine into ornithine and urea. This reaction is part of the urea cycle, which helps to rid the body of excess nitrogen waste produced during the metabolism of proteins. Arginase is found in various tissues throughout the body, including the liver, where it plays a key role in the detoxification of ammonia.

Cholesterol is a type of lipid (fat) molecule that is an essential component of cell membranes and is also used to make certain hormones and vitamins in the body. It is produced by the liver and is also obtained from animal-derived foods such as meat, dairy products, and eggs.

Cholesterol does not mix with blood, so it is transported through the bloodstream by lipoproteins, which are particles made up of both lipids and proteins. There are two main types of lipoproteins that carry cholesterol: low-density lipoproteins (LDL), also known as "bad" cholesterol, and high-density lipoproteins (HDL), also known as "good" cholesterol.

High levels of LDL cholesterol in the blood can lead to a buildup of cholesterol in the walls of the arteries, increasing the risk of heart disease and stroke. On the other hand, high levels of HDL cholesterol are associated with a lower risk of these conditions because HDL helps remove LDL cholesterol from the bloodstream and transport it back to the liver for disposal.

It is important to maintain healthy levels of cholesterol through a balanced diet, regular exercise, and sometimes medication if necessary. Regular screening is also recommended to monitor cholesterol levels and prevent health complications.

Fc receptors (FcRs) are specialized proteins found on the surface of various immune cells, including neutrophils, monocytes, macrophages, eosinophils, basophils, mast cells, and B lymphocytes. They play a crucial role in the immune response by recognizing and binding to the Fc region of antibodies (IgG, IgA, and IgE) after they have interacted with their specific antigens.

FcRs can be classified into several types based on the class of antibody they bind:

1. FcγRs - bind to the Fc region of IgG antibodies
2. FcαRs - bind to the Fc region of IgA antibodies
3. FcεRs - bind to the Fc region of IgE antibodies

The binding of antibodies to Fc receptors triggers various cellular responses, such as phagocytosis, degranulation, and antibody-dependent cellular cytotoxicity (ADCC), which contribute to the elimination of pathogens, immune complexes, and other foreign substances. Dysregulation of Fc receptor function has been implicated in several diseases, including autoimmune disorders and allergies.

Cell movement, also known as cell motility, refers to the ability of cells to move independently and change their location within tissue or inside the body. This process is essential for various biological functions, including embryonic development, wound healing, immune responses, and cancer metastasis.

There are several types of cell movement, including:

1. **Crawling or mesenchymal migration:** Cells move by extending and retracting protrusions called pseudopodia or filopodia, which contain actin filaments. This type of movement is common in fibroblasts, immune cells, and cancer cells during tissue invasion and metastasis.
2. **Amoeboid migration:** Cells move by changing their shape and squeezing through tight spaces without forming protrusions. This type of movement is often observed in white blood cells (leukocytes) as they migrate through the body to fight infections.
3. **Pseudopodial extension:** Cells extend pseudopodia, which are temporary cytoplasmic projections containing actin filaments. These protrusions help the cell explore its environment and move forward.
4. **Bacterial flagellar motion:** Bacteria use a whip-like structure called a flagellum to propel themselves through their environment. The rotation of the flagellum is driven by a molecular motor in the bacterial cell membrane.
5. **Ciliary and ependymal movement:** Ciliated cells, such as those lining the respiratory tract and fallopian tubes, have hair-like structures called cilia that beat in coordinated waves to move fluids or mucus across the cell surface.

Cell movement is regulated by a complex interplay of signaling pathways, cytoskeletal rearrangements, and adhesion molecules, which enable cells to respond to environmental cues and navigate through tissues.

C-type lectins are a family of proteins that contain one or more carbohydrate recognition domains (CRDs) with a characteristic pattern of conserved sequence motifs. These proteins are capable of binding to specific carbohydrate structures in a calcium-dependent manner, making them important in various biological processes such as cell adhesion, immune recognition, and initiation of inflammatory responses.

C-type lectins can be further classified into several subfamilies based on their structure and function, including selectins, collectins, and immunoglobulin-like receptors. They play a crucial role in the immune system by recognizing and binding to carbohydrate structures on the surface of pathogens, facilitating their clearance by phagocytic cells. Additionally, C-type lectins are involved in various physiological processes such as cell development, tissue repair, and cancer progression.

It is important to note that some C-type lectins can also bind to self-antigens and contribute to autoimmune diseases. Therefore, understanding the structure and function of these proteins has important implications for developing new therapeutic strategies for various diseases.

Toll-like receptor 2 (TLR2) is a type of protein belonging to the family of pattern recognition receptors (PRRs), which play a crucial role in the innate immune system's response to pathogens. TLR2 is primarily expressed on the surface of various immune cells, including monocytes, macrophages, dendritic cells, and B cells.

TLR2 recognizes a wide range of microbial components, such as lipopeptides, lipoteichoic acid, and zymosan, derived from both gram-positive and gram-negative bacteria, fungi, and certain viruses. Upon recognition and binding to these ligands, TLR2 initiates a signaling cascade that activates various transcription factors, leading to the production of proinflammatory cytokines, chemokines, and costimulatory molecules. This response is essential for the activation and recruitment of immune cells to the site of infection, thereby contributing to the clearance of invading pathogens.

In summary, TLR2 is a vital pattern recognition receptor that helps the innate immune system detect and respond to various microbial threats by initiating an inflammatory response upon ligand binding.

Interleukin-1 beta (IL-1β) is a member of the interleukin-1 cytokine family and is primarily produced by activated macrophages in response to inflammatory stimuli. It is a crucial mediator of the innate immune response and plays a key role in the regulation of various biological processes, including cell proliferation, differentiation, and apoptosis. IL-1β is involved in the pathogenesis of several inflammatory diseases, such as rheumatoid arthritis, inflammatory bowel disease, and atherosclerosis. It exerts its effects by binding to the interleukin-1 receptor, which triggers a signaling cascade that leads to the activation of various transcription factors and the expression of target genes.

"Cell count" is a medical term that refers to the process of determining the number of cells present in a given volume or sample of fluid or tissue. This can be done through various laboratory methods, such as counting individual cells under a microscope using a specialized grid called a hemocytometer, or using automated cell counters that use light scattering and electrical impedance techniques to count and classify different types of cells.

Cell counts are used in a variety of medical contexts, including hematology (the study of blood and blood-forming tissues), microbiology (the study of microscopic organisms), and pathology (the study of diseases and their causes). For example, a complete blood count (CBC) is a routine laboratory test that includes a white blood cell (WBC) count, red blood cell (RBC) count, hemoglobin level, hematocrit value, and platelet count. Abnormal cell counts can indicate the presence of various medical conditions, such as infections, anemia, or leukemia.

"Mycobacterium bovis" is a species of slow-growing, aerobic, gram-positive bacteria in the family Mycobacteriaceae. It is the causative agent of tuberculosis in cattle and other animals, and can also cause tuberculosis in humans, particularly in those who come into contact with infected animals or consume unpasteurized dairy products from infected cows. The bacteria are resistant to many common disinfectants and survive for long periods in a dormant state, making them difficult to eradicate from the environment. "Mycobacterium bovis" is closely related to "Mycobacterium tuberculosis," the bacterium that causes tuberculosis in humans, and both species share many genetic and biochemical characteristics.

Monokines are cytokines that are produced and released by monocytes, which are a type of white blood cell. These proteins play an important role in the immune response, including inflammation, immunoregulation, and hematopoiesis (the formation of blood cells).

Monokines include several types of cytokines, such as tumor necrosis factor-alpha (TNF-α), interleukin-1 (IL-1), interleukin-6 (IL-6), and interleukin-12 (IL-12). These molecules help to regulate the activity of other immune cells, such as T cells and B cells, and can also have direct effects on infected or damaged tissues.

Monokines are involved in a variety of physiological and pathological processes, including host defense against infection, tissue repair and regeneration, and the development of chronic inflammatory diseases such as rheumatoid arthritis and atherosclerosis.

Opsonins are proteins found in the blood that help enhance the immune system's response to foreign substances, such as bacteria and viruses. They do this by coating the surface of these pathogens, making them more recognizable to immune cells like neutrophils and macrophages. This process, known as opsonization, facilitates the phagocytosis (engulfing and destroying) of the pathogen by these immune cells.

There are two main types of opsonins:

1. IgG antibodies: These are a type of antibody produced by the immune system in response to an infection. They bind to specific antigens on the surface of the pathogen, marking them for destruction by phagocytic cells.
2. Complement proteins: The complement system is a group of proteins that work together to help eliminate pathogens. When activated, the complement system can produce various proteins that act as opsonins, including C3b and C4b. These proteins bind to the surface of the pathogen, making it easier for phagocytic cells to recognize and destroy them.

In summary, opsonin proteins are crucial components of the immune system's response to infections, helping to mark foreign substances for destruction by immune cells like neutrophils and macrophages.

Pinocytosis is a type of cellular process involving the ingestion and absorption of extracellular fluid and dissolved substances into a cell. It is a form of endocytosis, where the cell membrane surrounds and engulfs the extracellular fluid to form a vesicle containing the fluid and its contents within the cell cytoplasm.

In pinocytosis, the cell membrane invaginates and forms small vesicles (pinocytotic vesicles) that contain extracellular fluid and dissolved substances. These vesicles then detach from the cell membrane and move into the cytoplasm, where they fuse with endosomes or lysosomes to break down and digest the contents of the vesicle.

Pinocytosis is a non-selective process that allows cells to take up small amounts of extracellular fluid and dissolved substances from their environment. It plays an important role in various physiological processes, including nutrient uptake, cell signaling, and the regulation of extracellular matrix composition.

'Mycobacterium tuberculosis' is a species of slow-growing, aerobic, gram-positive bacteria that demonstrates acid-fastness. It is the primary causative agent of tuberculosis (TB) in humans. This bacterium has a complex cell wall rich in lipids, including mycolic acids, which provides a hydrophobic barrier and makes it resistant to many conventional antibiotics. The ability of M. tuberculosis to survive within host macrophages and resist the immune response contributes to its pathogenicity and the difficulty in treating TB infections.

M. tuberculosis is typically transmitted through inhalation of infectious droplets containing the bacteria, which primarily targets the lungs but can spread to other parts of the body (extrapulmonary TB). The infection may result in a spectrum of clinical manifestations, ranging from latent TB infection (LTBI) to active disease. LTBI represents a dormant state where individuals are infected with M. tuberculosis but do not show symptoms and cannot transmit the bacteria. However, they remain at risk of developing active TB throughout their lifetime, especially if their immune system becomes compromised.

Effective prevention and control strategies for TB rely on early detection, treatment, and public health interventions to limit transmission. The current first-line treatments for drug-susceptible TB include a combination of isoniazid, rifampin, ethambutol, and pyrazinamide for at least six months. Multidrug-resistant (MDR) and extensively drug-resistant (XDR) strains of M. tuberculosis present significant challenges in TB control and require more complex treatment regimens.

Membrane glycoproteins are proteins that contain oligosaccharide chains (glycans) covalently attached to their polypeptide backbone. They are integral components of biological membranes, spanning the lipid bilayer and playing crucial roles in various cellular processes.

The glycosylation of these proteins occurs in the endoplasmic reticulum (ER) and Golgi apparatus during protein folding and trafficking. The attached glycans can vary in structure, length, and composition, which contributes to the diversity of membrane glycoproteins.

Membrane glycoproteins can be classified into two main types based on their orientation within the lipid bilayer:

1. Type I (N-linked): These glycoproteins have a single transmembrane domain and an extracellular N-terminus, where the oligosaccharides are predominantly attached via asparagine residues (Asn-X-Ser/Thr sequon).
2. Type II (C-linked): These glycoproteins possess two transmembrane domains and an intracellular C-terminus, with the oligosaccharides linked to tryptophan residues via a mannose moiety.

Membrane glycoproteins are involved in various cellular functions, such as:

* Cell adhesion and recognition
* Receptor-mediated signal transduction
* Enzymatic catalysis
* Transport of molecules across membranes
* Cell-cell communication
* Immunological responses

Some examples of membrane glycoproteins include cell surface receptors (e.g., growth factor receptors, cytokine receptors), adhesion molecules (e.g., integrins, cadherins), and transporters (e.g., ion channels, ABC transporters).

Arteriosclerosis is a general term that describes the hardening and stiffening of the artery walls. It's a progressive condition that can occur as a result of aging, or it may be associated with certain risk factors such as high blood pressure, high cholesterol, diabetes, smoking, and a sedentary lifestyle.

The process of arteriosclerosis involves the buildup of plaque, made up of fat, cholesterol, calcium, and other substances, in the inner lining of the artery walls. Over time, this buildup can cause the artery walls to thicken and harden, reducing the flow of oxygen-rich blood to the body's organs and tissues.

Arteriosclerosis can affect any of the body's arteries, but it is most commonly found in the coronary arteries that supply blood to the heart, the cerebral arteries that supply blood to the brain, and the peripheral arteries that supply blood to the limbs. When arteriosclerosis affects the coronary arteries, it can lead to heart disease, angina, or heart attack. When it affects the cerebral arteries, it can lead to stroke or transient ischemic attack (TIA). When it affects the peripheral arteries, it can cause pain, numbness, or weakness in the limbs, and in severe cases, gangrene and amputation.

Apolipoprotein E (ApoE) is a protein involved in the metabolism of lipids, particularly cholesterol. It is produced primarily by the liver and is a component of several types of lipoproteins, including very low-density lipoproteins (VLDL) and high-density lipoproteins (HDL).

ApoE plays a crucial role in the transport and uptake of lipids in the body. It binds to specific receptors on cell surfaces, facilitating the delivery of lipids to cells for energy metabolism or storage. ApoE also helps to clear cholesterol from the bloodstream and is involved in the repair and maintenance of tissues.

There are three major isoforms of ApoE, designated ApoE2, ApoE3, and ApoE4, which differ from each other by only a few amino acids. These genetic variations can have significant effects on an individual's risk for developing certain diseases, particularly cardiovascular disease and Alzheimer's disease. For example, individuals who inherit the ApoE4 allele have an increased risk of developing Alzheimer's disease, while those with the ApoE2 allele may have a reduced risk.

In summary, Apolipoprotein E is a protein involved in lipid metabolism and transport, and genetic variations in this protein can influence an individual's risk for certain diseases.

IgG receptors, also known as Fcγ receptors (Fc gamma receptors), are specialized protein molecules found on the surface of various immune cells, such as neutrophils, monocytes, macrophages, and some lymphocytes. These receptors recognize and bind to the Fc region of IgG antibodies, one of the five classes of immunoglobulins in the human body.

IgG receptors play a crucial role in immune responses by mediating different effector functions, including:

1. Antibody-dependent cellular cytotoxicity (ADCC): IgG receptors on natural killer (NK) cells and other immune cells bind to IgG antibodies coated on the surface of virus-infected or cancer cells, leading to their destruction.
2. Phagocytosis: When IgG antibodies tag pathogens or foreign particles, phagocytes like neutrophils and macrophages recognize and bind to these immune complexes via IgG receptors, facilitating the engulfment and removal of the targeted particles.
3. Antigen presentation: IgG receptors on antigen-presenting cells (APCs) can internalize immune complexes, process the antigens, and present them to T cells, thereby initiating adaptive immune responses.
4. Inflammatory response regulation: IgG receptors can modulate inflammation by activating or inhibiting downstream signaling pathways in immune cells, depending on the specific type of Fcγ receptor and its activation state.

There are several types of IgG receptors (FcγRI, FcγRII, FcγRIII, and FcγRIV) with varying affinities for different subclasses of IgG antibodies (IgG1, IgG2, IgG3, and IgG4). The distinct functions and expression patterns of these receptors contribute to the complexity and fine-tuning of immune responses in the human body.

Molecular sequence data refers to the specific arrangement of molecules, most commonly nucleotides in DNA or RNA, or amino acids in proteins, that make up a biological macromolecule. This data is generated through laboratory techniques such as sequencing, and provides information about the exact order of the constituent molecules. This data is crucial in various fields of biology, including genetics, evolution, and molecular biology, allowing for comparisons between different organisms, identification of genetic variations, and studies of gene function and regulation.

Dinoprostone is a prostaglandin E2 analog used in medical practice for the induction of labor and ripening of the cervix in pregnant women. It is available in various forms, including vaginal suppositories, gel, and tablets. Dinoprostone works by stimulating the contraction of uterine muscles and promoting cervical dilation, which helps in facilitating a successful delivery.

It's important to note that dinoprostone should only be administered under the supervision of a healthcare professional, as its use is associated with certain risks and side effects, including uterine hyperstimulation, fetal distress, and maternal infection. The dosage and duration of treatment are carefully monitored to minimize these risks and ensure the safety of both the mother and the baby.

Cell survival refers to the ability of a cell to continue living and functioning normally, despite being exposed to potentially harmful conditions or treatments. This can include exposure to toxins, radiation, chemotherapeutic drugs, or other stressors that can damage cells or interfere with their normal processes.

In scientific research, measures of cell survival are often used to evaluate the effectiveness of various therapies or treatments. For example, researchers may expose cells to a particular drug or treatment and then measure the percentage of cells that survive to assess its potential therapeutic value. Similarly, in toxicology studies, measures of cell survival can help to determine the safety of various chemicals or substances.

It's important to note that cell survival is not the same as cell proliferation, which refers to the ability of cells to divide and multiply. While some treatments may promote cell survival, they may also inhibit cell proliferation, making them useful for treating diseases such as cancer. Conversely, other treatments may be designed to specifically target and kill cancer cells, even if it means sacrificing some healthy cells in the process.

Nitric Oxide Synthase (NOS) is a group of enzymes that catalyze the production of nitric oxide (NO) from L-arginine. There are three distinct isoforms of NOS, each with different expression patterns and functions:

1. Neuronal Nitric Oxide Synthase (nNOS or NOS1): This isoform is primarily expressed in the nervous system and plays a role in neurotransmission, synaptic plasticity, and learning and memory processes.
2. Inducible Nitric Oxide Synthase (iNOS or NOS2): This isoform is induced by various stimuli such as cytokines, lipopolysaccharides, and hypoxia in a variety of cells including immune cells, endothelial cells, and smooth muscle cells. iNOS produces large amounts of NO, which functions as a potent effector molecule in the immune response, particularly in the defense against microbial pathogens.
3. Endothelial Nitric Oxide Synthase (eNOS or NOS3): This isoform is constitutively expressed in endothelial cells and produces low levels of NO that play a crucial role in maintaining vascular homeostasis by regulating vasodilation, inhibiting platelet aggregation, and preventing smooth muscle cell proliferation.

Overall, NOS plays an essential role in various physiological processes, including neurotransmission, immune response, cardiovascular function, and respiratory regulation. Dysregulation of NOS activity has been implicated in several pathological conditions such as hypertension, atherosclerosis, neurodegenerative diseases, and inflammatory disorders.

Dendritic cells (DCs) are a type of immune cell that play a critical role in the body's defense against infection and cancer. They are named for their dendrite-like projections, which they use to interact with and sample their environment. DCs are responsible for processing antigens (foreign substances that trigger an immune response) and presenting them to T cells, a type of white blood cell that plays a central role in the immune system's response to infection and cancer.

DCs can be found throughout the body, including in the skin, mucous membranes, and lymphoid organs. They are able to recognize and respond to a wide variety of antigens, including those from bacteria, viruses, fungi, and parasites. Once they have processed an antigen, DCs migrate to the lymph nodes, where they present the antigen to T cells. This interaction activates the T cells, which then go on to mount a targeted immune response against the invading pathogen or cancerous cells.

DCs are a diverse group of cells that can be divided into several subsets based on their surface markers and function. Some DCs, such as Langerhans cells and dermal DCs, are found in the skin and mucous membranes, where they serve as sentinels for invading pathogens. Other DCs, such as plasmacytoid DCs and conventional DCs, are found in the lymphoid organs, where they play a role in activating T cells and initiating an immune response.

Overall, dendritic cells are essential for the proper functioning of the immune system, and dysregulation of these cells has been implicated in a variety of diseases, including autoimmune disorders and cancer.

Coculture techniques refer to a type of experimental setup in which two or more different types of cells or organisms are grown and studied together in a shared culture medium. This method allows researchers to examine the interactions between different cell types or species under controlled conditions, and to study how these interactions may influence various biological processes such as growth, gene expression, metabolism, and signal transduction.

Coculture techniques can be used to investigate a wide range of biological phenomena, including the effects of host-microbe interactions on human health and disease, the impact of different cell types on tissue development and homeostasis, and the role of microbial communities in shaping ecosystems. These techniques can also be used to test the efficacy and safety of new drugs or therapies by examining their effects on cells grown in coculture with other relevant cell types.

There are several different ways to establish cocultures, depending on the specific research question and experimental goals. Some common methods include:

1. Mixed cultures: In this approach, two or more cell types are simply mixed together in a culture dish or flask and allowed to grow and interact freely.
2. Cell-layer cultures: Here, one cell type is grown on a porous membrane or other support structure, while the second cell type is grown on top of it, forming a layered coculture.
3. Conditioned media cultures: In this case, one cell type is grown to confluence and its culture medium is collected and then used to grow a second cell type. This allows the second cell type to be exposed to any factors secreted by the first cell type into the medium.
4. Microfluidic cocultures: These involve growing cells in microfabricated channels or chambers, which allow for precise control over the spatial arrangement and flow of nutrients, waste products, and signaling molecules between different cell types.

Overall, coculture techniques provide a powerful tool for studying complex biological systems and gaining insights into the mechanisms that underlie various physiological and pathological processes.

Lymphocytes are a type of white blood cell that is an essential part of the immune system. They are responsible for recognizing and responding to potentially harmful substances such as viruses, bacteria, and other foreign invaders. There are two main types of lymphocytes: B-lymphocytes (B-cells) and T-lymphocytes (T-cells).

B-lymphocytes produce antibodies, which are proteins that help to neutralize or destroy foreign substances. When a B-cell encounters a foreign substance, it becomes activated and begins to divide and differentiate into plasma cells, which produce and secrete large amounts of antibodies. These antibodies bind to the foreign substance, marking it for destruction by other immune cells.

T-lymphocytes, on the other hand, are involved in cell-mediated immunity. They directly attack and destroy infected cells or cancerous cells. T-cells can also help to regulate the immune response by producing chemical signals that activate or inhibit other immune cells.

Lymphocytes are produced in the bone marrow and mature in either the bone marrow (B-cells) or the thymus gland (T-cells). They circulate throughout the body in the blood and lymphatic system, where they can be found in high concentrations in lymph nodes, the spleen, and other lymphoid organs.

Abnormalities in the number or function of lymphocytes can lead to a variety of immune-related disorders, including immunodeficiency diseases, autoimmune disorders, and cancer.

Toll-like receptors (TLRs) are a type of pattern recognition receptors (PRRs) that play a crucial role in the innate immune system. They are transmembrane proteins located on the surface of various immune cells, including macrophages, dendritic cells, and B cells. TLRs recognize specific patterns of molecules called pathogen-associated molecular patterns (PAMPs) that are found on microbes such as bacteria, viruses, fungi, and parasites.

Once TLRs bind to PAMPs, they initiate a signaling cascade that activates the immune response, leading to the production of cytokines and chemokines, which in turn recruit and activate other immune cells. TLRs also play a role in the adaptive immune response by activating antigen-presenting cells and promoting the differentiation of T cells.

There are ten known human TLRs, each with distinct ligand specificity and cellular localization. TLRs can be found on the cell surface or within endosomes, where they recognize different types of PAMPs. For example, TLR4 recognizes lipopolysaccharides (LPS) found on gram-negative bacteria, while TLR3 recognizes double-stranded RNA from viruses.

Overall, TLRs are critical components of the immune system's ability to detect and respond to infections, and dysregulation of TLR signaling has been implicated in various inflammatory diseases and cancers.

Western blotting is a laboratory technique used in molecular biology to detect and quantify specific proteins in a mixture of many different proteins. This technique is commonly used to confirm the expression of a protein of interest, determine its size, and investigate its post-translational modifications. The name "Western" blotting distinguishes this technique from Southern blotting (for DNA) and Northern blotting (for RNA).

The Western blotting procedure involves several steps:

1. Protein extraction: The sample containing the proteins of interest is first extracted, often by breaking open cells or tissues and using a buffer to extract the proteins.
2. Separation of proteins by electrophoresis: The extracted proteins are then separated based on their size by loading them onto a polyacrylamide gel and running an electric current through the gel (a process called sodium dodecyl sulfate-polyacrylamide gel electrophoresis or SDS-PAGE). This separates the proteins according to their molecular weight, with smaller proteins migrating faster than larger ones.
3. Transfer of proteins to a membrane: After separation, the proteins are transferred from the gel onto a nitrocellulose or polyvinylidene fluoride (PVDF) membrane using an electric current in a process called blotting. This creates a replica of the protein pattern on the gel but now immobilized on the membrane for further analysis.
4. Blocking: The membrane is then blocked with a blocking agent, such as non-fat dry milk or bovine serum albumin (BSA), to prevent non-specific binding of antibodies in subsequent steps.
5. Primary antibody incubation: A primary antibody that specifically recognizes the protein of interest is added and allowed to bind to its target protein on the membrane. This step may be performed at room temperature or 4°C overnight, depending on the antibody's properties.
6. Washing: The membrane is washed with a buffer to remove unbound primary antibodies.
7. Secondary antibody incubation: A secondary antibody that recognizes the primary antibody (often coupled to an enzyme or fluorophore) is added and allowed to bind to the primary antibody. This step may involve using a horseradish peroxidase (HRP)-conjugated or alkaline phosphatase (AP)-conjugated secondary antibody, depending on the detection method used later.
8. Washing: The membrane is washed again to remove unbound secondary antibodies.
9. Detection: A detection reagent is added to visualize the protein of interest by detecting the signal generated from the enzyme-conjugated or fluorophore-conjugated secondary antibody. This can be done using chemiluminescent, colorimetric, or fluorescent methods.
10. Analysis: The resulting image is analyzed to determine the presence and quantity of the protein of interest in the sample.

Western blotting is a powerful technique for identifying and quantifying specific proteins within complex mixtures. It can be used to study protein expression, post-translational modifications, protein-protein interactions, and more. However, it requires careful optimization and validation to ensure accurate and reproducible results.

ATP Binding Cassette Transporter 1 (ABC Transporter 1 or ABCB1) is a protein that belongs to the superfamily of ATP-binding cassette (ABC) transporters. These proteins utilize the energy from ATP hydrolysis to transport various substrates across membranes.

The ABCB1 gene encodes for the P-glycoprotein (P-gp), a 170 kDa protein, which is an efflux transporter primarily located in the plasma membrane of various cell types, including epithelial and endothelial cells. P-gp plays a crucial role in limiting the absorption and facilitating the excretion of many drugs by actively pumping them out of cells, thereby contributing to multidrug resistance (MDR) in cancer cells.

P-gp has a broad substrate specificity and can transport various structurally diverse compounds, including chemotherapeutic agents, antibiotics, antiviral drugs, and natural toxins. Its expression is often upregulated in cancer cells, leading to reduced intracellular drug accumulation and decreased therapeutic efficacy. In addition to its role in drug resistance, P-gp also functions in the absorption, distribution, and excretion of drugs in normal tissues, particularly in the intestine, liver, and kidney.

Chemokine (C-X-C motif) ligand 2, also known as CXCL2, is a small signaling protein that belongs to the chemokine family. Chemokines are a group of cytokines, or cell signaling molecules, that play crucial roles in immune responses and inflammation. They mediate their effects by interacting with specific receptors on the surface of target cells, guiding the migration of various immune cells to sites of infection, injury, or inflammation.

CXCL2 is primarily produced by activated monocytes, macrophages, and neutrophils, as well as endothelial cells, fibroblasts, and certain types of tumor cells. Its primary function is to attract and activate neutrophils, which are key effector cells in the early stages of inflammation and host defense against invading pathogens. CXCL2 exerts its effects by binding to its specific receptor, CXCR2, which is expressed on the surface of neutrophils and other immune cells.

In addition to its role in inflammation and immunity, CXCL2 has been implicated in various pathological conditions, including cancer, atherosclerosis, and autoimmune diseases. Its expression can be regulated by several factors, such as pro-inflammatory cytokines, bacterial products, and growth factors. Understanding the role of CXCL2 in health and disease may provide insights into the development of novel therapeutic strategies for treating inflammation-associated disorders.

Transgenic mice are genetically modified rodents that have incorporated foreign DNA (exogenous DNA) into their own genome. This is typically done through the use of recombinant DNA technology, where a specific gene or genetic sequence of interest is isolated and then introduced into the mouse embryo. The resulting transgenic mice can then express the protein encoded by the foreign gene, allowing researchers to study its function in a living organism.

The process of creating transgenic mice usually involves microinjecting the exogenous DNA into the pronucleus of a fertilized egg, which is then implanted into a surrogate mother. The offspring that result from this procedure are screened for the presence of the foreign DNA, and those that carry the desired genetic modification are used to establish a transgenic mouse line.

Transgenic mice have been widely used in biomedical research to model human diseases, study gene function, and test new therapies. They provide a valuable tool for understanding complex biological processes and developing new treatments for a variety of medical conditions.

Electron microscopy (EM) is a type of microscopy that uses a beam of electrons to create an image of the sample being examined, resulting in much higher magnification and resolution than light microscopy. There are several types of electron microscopy, including transmission electron microscopy (TEM), scanning electron microscopy (SEM), and reflection electron microscopy (REM).

In TEM, a beam of electrons is transmitted through a thin slice of the sample, and the electrons that pass through the sample are focused to form an image. This technique can provide detailed information about the internal structure of cells, viruses, and other biological specimens, as well as the composition and structure of materials at the atomic level.

In SEM, a beam of electrons is scanned across the surface of the sample, and the electrons that are scattered back from the surface are detected to create an image. This technique can provide information about the topography and composition of surfaces, as well as the structure of materials at the microscopic level.

REM is a variation of SEM in which the beam of electrons is reflected off the surface of the sample, rather than scattered back from it. This technique can provide information about the surface chemistry and composition of materials.

Electron microscopy has a wide range of applications in biology, medicine, and materials science, including the study of cellular structure and function, disease diagnosis, and the development of new materials and technologies.

Superoxides are partially reduced derivatives of oxygen that contain one extra electron, giving them an overall charge of -1. They are highly reactive and unstable, with the most common superoxide being the hydroxyl radical (•OH-) and the superoxide anion (O2-). Superoxides are produced naturally in the body during metabolic processes, particularly within the mitochondria during cellular respiration. They play a role in various physiological processes, but when produced in excess or not properly neutralized, they can contribute to oxidative stress and damage to cells and tissues, potentially leading to the development of various diseases such as cancer, atherosclerosis, and neurodegenerative disorders.

The Macrophage-1 Antigen (also known as Macrophage Antigen-1 or CD14) is a glycoprotein found on the surface of various cells, including monocytes, macrophages, and some dendritic cells. It functions as a receptor for complexes formed by lipopolysaccharides (LPS) and LPS-binding protein (LBP), which are involved in the immune response to gram-negative bacteria. CD14 plays a crucial role in activating immune cells and initiating the release of proinflammatory cytokines upon recognizing bacterial components.

In summary, Macrophage-1 Antigen is a cell surface receptor that contributes to the recognition and response against gram-negative bacteria by interacting with LPS-LBP complexes.

Interleukin-12 (IL-12) is a naturally occurring protein that is primarily produced by activated macrophages and dendritic cells, which are types of immune cells. It plays a crucial role in the regulation of the immune response, particularly in the development of cell-mediated immunity.

IL-12 is composed of two subunits, p35 and p40, which combine to form a heterodimer. This cytokine stimulates the differentiation and activation of naive T cells into Th1 cells, which are important for fighting intracellular pathogens such as viruses and bacteria. IL-12 also enhances the cytotoxic activity of natural killer (NK) cells and CD8+ T cells, which can directly kill infected or malignant cells.

In addition to its role in the immune response, IL-12 has been implicated in the pathogenesis of several autoimmune diseases, including multiple sclerosis, rheumatoid arthritis, and psoriasis. As a result, therapeutic strategies targeting IL-12 or its signaling pathways have been explored as potential treatments for these conditions.

"Listeria monocytogenes" is a gram-positive, facultatively anaerobic, rod-shaped bacterium that is a major cause of foodborne illness. It is widely distributed in the environment and can be found in water, soil, vegetation, and various animal species. This pathogen is particularly notable for its ability to grow at low temperatures, allowing it to survive and multiply in refrigerated foods.

In humans, Listeria monocytogenes can cause a serious infection known as listeriosis, which primarily affects pregnant women, newborns, older adults, and individuals with weakened immune systems. The bacterium can cross the intestinal barrier, enter the bloodstream, and spread to the central nervous system, causing meningitis or encephalitis. Pregnant women infected with Listeria monocytogenes may experience mild flu-like symptoms but are at risk of transmitting the infection to their unborn children, which can result in stillbirth, premature delivery, or severe illness in newborns.

Common sources of Listeria monocytogenes include raw or undercooked meat, poultry, and seafood; unpasteurized dairy products; and ready-to-eat foods like deli meats, hot dogs, and soft cheeses. Proper food handling, cooking, and storage practices can help prevent listeriosis.

Kupffer cells are specialized macrophages that reside in the liver, particularly in the sinusoids of the liver's blood circulation system. They play a crucial role in the immune system by engulfing and destroying bacteria, microorganisms, and other particles that enter the liver via the portal vein. Kupffer cells also contribute to the clearance of damaged red blood cells, iron metabolism, and the regulation of inflammation in the liver. They are named after the German pathologist Karl Wilhelm von Kupffer who first described them in 1876.

Monoclonal antibodies are a type of antibody that are identical because they are produced by a single clone of cells. They are laboratory-produced molecules that act like human antibodies in the immune system. They can be designed to attach to specific proteins found on the surface of cancer cells, making them useful for targeting and treating cancer. Monoclonal antibodies can also be used as a therapy for other diseases, such as autoimmune disorders and inflammatory conditions.

Monoclonal antibodies are produced by fusing a single type of immune cell, called a B cell, with a tumor cell to create a hybrid cell, or hybridoma. This hybrid cell is then able to replicate indefinitely, producing a large number of identical copies of the original antibody. These antibodies can be further modified and engineered to enhance their ability to bind to specific targets, increase their stability, and improve their effectiveness as therapeutic agents.

Monoclonal antibodies have several mechanisms of action in cancer therapy. They can directly kill cancer cells by binding to them and triggering an immune response. They can also block the signals that promote cancer growth and survival. Additionally, monoclonal antibodies can be used to deliver drugs or radiation directly to cancer cells, increasing the effectiveness of these treatments while minimizing their side effects on healthy tissues.

Monoclonal antibodies have become an important tool in modern medicine, with several approved for use in cancer therapy and other diseases. They are continuing to be studied and developed as a promising approach to treating a wide range of medical conditions.

A dose-response relationship in the context of drugs refers to the changes in the effects or symptoms that occur as the dose of a drug is increased or decreased. Generally, as the dose of a drug is increased, the severity or intensity of its effects also increases. Conversely, as the dose is decreased, the effects of the drug become less severe or may disappear altogether.

The dose-response relationship is an important concept in pharmacology and toxicology because it helps to establish the safe and effective dosage range for a drug. By understanding how changes in the dose of a drug affect its therapeutic and adverse effects, healthcare providers can optimize treatment plans for their patients while minimizing the risk of harm.

The dose-response relationship is typically depicted as a curve that shows the relationship between the dose of a drug and its effect. The shape of the curve may vary depending on the drug and the specific effect being measured. Some drugs may have a steep dose-response curve, meaning that small changes in the dose can result in large differences in the effect. Other drugs may have a more gradual dose-response curve, where larger changes in the dose are needed to produce significant effects.

In addition to helping establish safe and effective dosages, the dose-response relationship is also used to evaluate the potential therapeutic benefits and risks of new drugs during clinical trials. By systematically testing different doses of a drug in controlled studies, researchers can identify the optimal dosage range for the drug and assess its safety and efficacy.

"Legionella pneumophila" is a species of Gram-negative, aerobic bacteria that are commonly found in freshwater environments such as lakes and streams. It can also be found in man-made water systems like hot tubs, cooling towers, and decorative fountains. This bacterium is the primary cause of Legionnaires' disease, a severe form of pneumonia, and Pontiac fever, a milder illness resembling the flu. Infection typically occurs when people inhale tiny droplets of water containing the bacteria. It is not transmitted from person to person.

Matrix metalloproteinase 12 (MMP-12) is a type of enzyme that belongs to the matrix metalloproteinase (MMP) family. MMPs are involved in the breakdown and remodeling of extracellular matrices, which are the structures that provide support and organization to cells in tissues and organs.

MMP-12 is also known as macrophage elastase because it is primarily produced by macrophages, a type of white blood cell that plays a key role in the immune system. MMP-12 is capable of degrading various components of the extracellular matrix, including elastin, a protein that provides elasticity to tissues such as lungs, arteries, and skin.

MMP-12 has been implicated in several physiological and pathological processes, including tissue remodeling, wound healing, inflammation, and cancer. Dysregulation of MMP-12 activity has been associated with various diseases, such as chronic obstructive pulmonary disease (COPD), atherosclerosis, and tumor metastasis.

"Mycobacterium avium is a species of gram-positive, aerobic bacteria that belongs to the family Mycobacteriaceae. It is a slow-growing mycobacterium that is widely distributed in the environment, particularly in soil and water. M. avium is an opportunistic pathogen that can cause pulmonary disease, lymphadenitis, and disseminated infection in individuals with compromised immune systems, such as those with HIV/AIDS. It is also known to cause pulmonary disease in elderly people with structural lung damage. The bacteria are resistant to many common disinfectants and can survive in hostile environments for extended periods."

Conditioned culture media refers to a type of growth medium that has been previously used to culture and maintain the cells of an organism. The conditioned media contains factors secreted by those cells, such as hormones, nutrients, and signaling molecules, which can affect the behavior and growth of other cells that are introduced into the media later on.

When the conditioned media is used for culturing a new set of cells, it can provide a more physiologically relevant environment than traditional culture media, as it contains factors that are specific to the original cell type. This can be particularly useful in studies that aim to understand cell-cell interactions and communication, or to mimic the natural microenvironment of cells in the body.

It's important to note that conditioned media should be handled carefully and used promptly after preparation, as the factors it contains can degrade over time and affect the quality of the results.

CD11b, also known as integrin αM or Mac-1, is not an antigen itself but a protein that forms part of a family of cell surface receptors called integrins. These integrins play a crucial role in various biological processes, including cell adhesion, migration, and signaling.

CD11b combines with CD18 (integrin β2) to form the heterodimeric integrin αMβ2, also known as Mac-1 or CR3 (complement receptor 3). This integrin is primarily expressed on the surface of myeloid cells, such as monocytes, macrophages, and neutrophils.

As an integral part of the immune system, CD11b/CD18 recognizes and binds to various ligands, including:

1. Icosahedral bacterial components like lipopolysaccharides (LPS) and peptidoglycans
2. Fragments of complement component C3b (iC3b)
3. Fibrinogen and other extracellular matrix proteins
4. Certain immune cell receptors, such as ICAM-1 (intercellular adhesion molecule 1)

The binding of CD11b/CD18 to these ligands triggers various intracellular signaling pathways that regulate the immune response and inflammation. In this context, antigens are substances (usually proteins or polysaccharides) found on the surface of cells, viruses, or bacteria that can be recognized by the immune system. CD11b/CD18 plays a role in recognizing and responding to these antigens during an immune response.

Cell adhesion refers to the binding of cells to extracellular matrices or to other cells, a process that is fundamental to the development, function, and maintenance of multicellular organisms. Cell adhesion is mediated by various cell surface receptors, such as integrins, cadherins, and immunoglobulin-like cell adhesion molecules (Ig-CAMs), which interact with specific ligands in the extracellular environment. These interactions lead to the formation of specialized junctions, such as tight junctions, adherens junctions, and desmosomes, that help to maintain tissue architecture and regulate various cellular processes, including proliferation, differentiation, migration, and survival. Disruptions in cell adhesion can contribute to a variety of diseases, including cancer, inflammation, and degenerative disorders.

Down-regulation is a process that occurs in response to various stimuli, where the number or sensitivity of cell surface receptors or the expression of specific genes is decreased. This process helps maintain homeostasis within cells and tissues by reducing the ability of cells to respond to certain signals or molecules.

In the context of cell surface receptors, down-regulation can occur through several mechanisms:

1. Receptor internalization: After binding to their ligands, receptors can be internalized into the cell through endocytosis. Once inside the cell, these receptors may be degraded or recycled back to the cell surface in smaller numbers.
2. Reduced receptor synthesis: Down-regulation can also occur at the transcriptional level, where the expression of genes encoding for specific receptors is decreased, leading to fewer receptors being produced.
3. Receptor desensitization: Prolonged exposure to a ligand can lead to a decrease in receptor sensitivity or affinity, making it more difficult for the cell to respond to the signal.

In the context of gene expression, down-regulation refers to the decreased transcription and/or stability of specific mRNAs, leading to reduced protein levels. This process can be induced by various factors, including microRNA (miRNA)-mediated regulation, histone modification, or DNA methylation.

Down-regulation is an essential mechanism in many physiological processes and can also contribute to the development of several diseases, such as cancer and neurodegenerative disorders.

U937 cells are a type of human histiocytic lymphoma cell line that is commonly used in scientific research and studies. They are derived from the peripheral blood of a patient with histiocytic lymphoma, which is a rare type of cancer that affects the immune system's cells called histiocytes.

U937 cells have a variety of uses in research, including studying the mechanisms of cancer cell growth and proliferation, testing the effects of various drugs and treatments on cancer cells, and investigating the role of different genes and proteins in cancer development and progression. These cells are easy to culture and maintain in the laboratory, making them a popular choice for researchers in many fields.

It is important to note that while U937 cells can provide valuable insights into the behavior of cancer cells, they do not necessarily reflect the complexity and diversity of human cancers. Therefore, findings from studies using these cells should be validated in more complex models or clinical trials before being applied to patient care.

I believe there may be some confusion in your question. "Rabbits" is a common name used to refer to the Lagomorpha species, particularly members of the family Leporidae. They are small mammals known for their long ears, strong legs, and quick reproduction.

However, if you're referring to "rabbits" in a medical context, there is a term called "rabbit syndrome," which is a rare movement disorder characterized by repetitive, involuntary movements of the fingers, resembling those of a rabbit chewing. It is also known as "finger-chewing chorea." This condition is usually associated with certain medications, particularly antipsychotics, and typically resolves when the medication is stopped or adjusted.

An Enzyme-Linked Immunosorbent Assay (ELISA) is a type of analytical biochemistry assay used to detect and quantify the presence of a substance, typically a protein or peptide, in a liquid sample. It takes its name from the enzyme-linked antibodies used in the assay.

In an ELISA, the sample is added to a well containing a surface that has been treated to capture the target substance. If the target substance is present in the sample, it will bind to the surface. Next, an enzyme-linked antibody specific to the target substance is added. This antibody will bind to the captured target substance if it is present. After washing away any unbound material, a substrate for the enzyme is added. If the enzyme is present due to its linkage to the antibody, it will catalyze a reaction that produces a detectable signal, such as a color change or fluorescence. The intensity of this signal is proportional to the amount of target substance present in the sample, allowing for quantification.

ELISAs are widely used in research and clinical settings to detect and measure various substances, including hormones, viruses, and bacteria. They offer high sensitivity, specificity, and reproducibility, making them a reliable choice for many applications.

CD36 is a type of protein found on the surface of certain cells in the human body, including platelets, white blood cells (monocytes and macrophages), and fat (adipose) cells. It is a type of scavenger receptor that plays a role in various biological processes, such as:

1. Fatty acid uptake and metabolism: CD36 helps facilitate the transport of long-chain fatty acids into cells for energy production and storage.
2. Inflammation and immune response: CD36 is involved in the recognition and clearance of foreign substances (pathogens) and damaged or dying cells, which can trigger an immune response.
3. Angiogenesis: CD36 has been implicated in the regulation of blood vessel formation (angiogenesis), particularly during wound healing and tumor growth.
4. Atherosclerosis: CD36 has been associated with the development and progression of atherosclerosis, a condition characterized by the buildup of fats, cholesterol, and other substances in and on the artery walls. This is due to its role in the uptake of oxidized low-density lipoprotein (oxLDL) by macrophages, leading to the formation of foam cells and the development of fatty streaks in the arterial wall.
5. Infectious diseases: CD36 has been identified as a receptor for various pathogens, including malaria parasites, HIV, and some bacteria, which can use this protein to gain entry into host cells.

As an antigen, CD36 is a molecule that can be targeted by the immune system to produce an immune response. Antibodies against CD36 have been found in various diseases, such as autoimmune disorders and certain infections. Modulation of CD36 activity has been suggested as a potential therapeutic strategy for several conditions, including atherosclerosis, diabetes, and infectious diseases.

Membrane proteins are a type of protein that are embedded in the lipid bilayer of biological membranes, such as the plasma membrane of cells or the inner membrane of mitochondria. These proteins play crucial roles in various cellular processes, including:

1. Cell-cell recognition and signaling
2. Transport of molecules across the membrane (selective permeability)
3. Enzymatic reactions at the membrane surface
4. Energy transduction and conversion
5. Mechanosensation and signal transduction

Membrane proteins can be classified into two main categories: integral membrane proteins, which are permanently associated with the lipid bilayer, and peripheral membrane proteins, which are temporarily or loosely attached to the membrane surface. Integral membrane proteins can further be divided into three subcategories based on their topology:

1. Transmembrane proteins, which span the entire width of the lipid bilayer with one or more alpha-helices or beta-barrels.
2. Lipid-anchored proteins, which are covalently attached to lipids in the membrane via a glycosylphosphatidylinositol (GPI) anchor or other lipid modifications.
3. Monotopic proteins, which are partially embedded in the membrane and have one or more domains exposed to either side of the bilayer.

Membrane proteins are essential for maintaining cellular homeostasis and are targets for various therapeutic interventions, including drug development and gene therapy. However, their structural complexity and hydrophobicity make them challenging to study using traditional biochemical methods, requiring specialized techniques such as X-ray crystallography, nuclear magnetic resonance (NMR) spectroscopy, and single-particle cryo-electron microscopy (cryo-EM).

Virus replication is the process by which a virus produces copies or reproduces itself inside a host cell. This involves several steps:

1. Attachment: The virus attaches to a specific receptor on the surface of the host cell.
2. Penetration: The viral genetic material enters the host cell, either by invagination of the cell membrane or endocytosis.
3. Uncoating: The viral genetic material is released from its protective coat (capsid) inside the host cell.
4. Replication: The viral genetic material uses the host cell's machinery to produce new viral components, such as proteins and nucleic acids.
5. Assembly: The newly synthesized viral components are assembled into new virus particles.
6. Release: The newly formed viruses are released from the host cell, often through lysis (breaking) of the cell membrane or by budding off the cell membrane.

The specific mechanisms and details of virus replication can vary depending on the type of virus. Some viruses, such as DNA viruses, use the host cell's DNA polymerase to replicate their genetic material, while others, such as RNA viruses, use their own RNA-dependent RNA polymerase or reverse transcriptase enzymes. Understanding the process of virus replication is important for developing antiviral therapies and vaccines.

Virulence, in the context of medicine and microbiology, refers to the degree or severity of damage or harm that a pathogen (like a bacterium, virus, fungus, or parasite) can cause to its host. It is often associated with the ability of the pathogen to invade and damage host tissues, evade or suppress the host's immune response, replicate within the host, and spread between hosts.

Virulence factors are the specific components or mechanisms that contribute to a pathogen's virulence, such as toxins, enzymes, adhesins, and capsules. These factors enable the pathogen to establish an infection, cause tissue damage, and facilitate its transmission between hosts. The overall virulence of a pathogen can be influenced by various factors, including host susceptibility, environmental conditions, and the specific strain or species of the pathogen.

A phenotype is the physical or biochemical expression of an organism's genes, or the observable traits and characteristics resulting from the interaction of its genetic constitution (genotype) with environmental factors. These characteristics can include appearance, development, behavior, and resistance to disease, among others. Phenotypes can vary widely, even among individuals with identical genotypes, due to differences in environmental influences, gene expression, and genetic interactions.

A base sequence in the context of molecular biology refers to the specific order of nucleotides in a DNA or RNA molecule. In DNA, these nucleotides are adenine (A), guanine (G), cytosine (C), and thymine (T). In RNA, uracil (U) takes the place of thymine. The base sequence contains genetic information that is transcribed into RNA and ultimately translated into proteins. It is the exact order of these bases that determines the genetic code and thus the function of the DNA or RNA molecule.

In the context of medicine and pharmacology, "kinetics" refers to the study of how a drug moves throughout the body, including its absorption, distribution, metabolism, and excretion (often abbreviated as ADME). This field is called "pharmacokinetics."

1. Absorption: This is the process of a drug moving from its site of administration into the bloodstream. Factors such as the route of administration (e.g., oral, intravenous, etc.), formulation, and individual physiological differences can affect absorption.

2. Distribution: Once a drug is in the bloodstream, it gets distributed throughout the body to various tissues and organs. This process is influenced by factors like blood flow, protein binding, and lipid solubility of the drug.

3. Metabolism: Drugs are often chemically modified in the body, typically in the liver, through processes known as metabolism. These changes can lead to the formation of active or inactive metabolites, which may then be further distributed, excreted, or undergo additional metabolic transformations.

4. Excretion: This is the process by which drugs and their metabolites are eliminated from the body, primarily through the kidneys (urine) and the liver (bile).

Understanding the kinetics of a drug is crucial for determining its optimal dosing regimen, potential interactions with other medications or foods, and any necessary adjustments for special populations like pediatric or geriatric patients, or those with impaired renal or hepatic function.

Immunologic cytotoxicity refers to the damage or destruction of cells that occurs as a result of an immune response. This process involves the activation of immune cells, such as cytotoxic T cells and natural killer (NK) cells, which release toxic substances, such as perforins and granzymes, that can kill target cells.

In addition, antibodies produced by B cells can also contribute to immunologic cytotoxicity by binding to antigens on the surface of target cells and triggering complement-mediated lysis or antibody-dependent cellular cytotoxicity (ADCC) by activating immune effector cells.

Immunologic cytotoxicity plays an important role in the body's defense against viral infections, cancer cells, and other foreign substances. However, it can also contribute to tissue damage and autoimmune diseases if the immune system mistakenly targets healthy cells or tissues.

Microglia are a type of specialized immune cell found in the brain and spinal cord. They are part of the glial family, which provide support and protection to the neurons in the central nervous system (CNS). Microglia account for about 10-15% of all cells found in the CNS.

The primary role of microglia is to constantly survey their environment and eliminate any potentially harmful agents, such as pathogens, dead cells, or protein aggregates. They do this through a process called phagocytosis, where they engulf and digest foreign particles or cellular debris. In addition to their phagocytic function, microglia also release various cytokines, chemokines, and growth factors that help regulate the immune response in the CNS, promote neuronal survival, and contribute to synaptic plasticity.

Microglia can exist in different activation states depending on the nature of the stimuli they encounter. In a resting state, microglia have a small cell body with numerous branches that are constantly monitoring their surroundings. When activated by an injury, infection, or neurodegenerative process, microglia change their morphology and phenotype, retracting their processes and adopting an amoeboid shape to migrate towards the site of damage or inflammation. Based on the type of activation, microglia can release both pro-inflammatory and anti-inflammatory factors that contribute to either neuroprotection or neurotoxicity.

Dysregulation of microglial function has been implicated in several neurological disorders, including Alzheimer's disease, Parkinson's disease, multiple sclerosis, and Amyotrophic Lateral Sclerosis (ALS). Therefore, understanding the role of microglia in health and disease is crucial for developing novel therapeutic strategies to treat these conditions.

A granuloma is a small, nodular inflammatory lesion that occurs in various tissues in response to chronic infection, foreign body reaction, or autoimmune conditions. Histologically, it is characterized by the presence of epithelioid macrophages, which are specialized immune cells with enlarged nuclei and abundant cytoplasm, often arranged in a palisading pattern around a central area containing necrotic debris, microorganisms, or foreign material.

Granulomas can be found in various medical conditions such as tuberculosis, sarcoidosis, fungal infections, and certain autoimmune disorders like Crohn's disease. The formation of granulomas is a complex process involving both innate and adaptive immune responses, which aim to contain and eliminate the offending agent while minimizing tissue damage.

Histocompatibility antigens Class II are a group of cell surface proteins that play a crucial role in the immune system's response to foreign substances. They are expressed on the surface of various cells, including immune cells such as B lymphocytes, macrophages, dendritic cells, and activated T lymphocytes.

Class II histocompatibility antigens are encoded by the major histocompatibility complex (MHC) class II genes, which are located on chromosome 6 in humans. These antigens are composed of two non-covalently associated polypeptide chains, an alpha (α) and a beta (β) chain, which form a heterodimer. There are three main types of Class II histocompatibility antigens, known as HLA-DP, HLA-DQ, and HLA-DR.

Class II histocompatibility antigens present peptide antigens to CD4+ T helper cells, which then activate other immune cells, such as B cells and macrophages, to mount an immune response against the presented antigen. Because of their role in initiating an immune response, Class II histocompatibility antigens are important in transplantation medicine, where mismatches between donor and recipient can lead to rejection of the transplanted organ or tissue.

A cell migration assay for macrophages is a type of experimental procedure used to study the movement and migration patterns of macrophages, a type of white blood cell that plays a crucial role in the immune system. These assays are often used in research to understand the mechanisms that control macrophage migration, which can be important in the context of various physiological and pathophysiological processes, such as wound healing, inflammation, and cancer metastasis.

There are several types of cell migration assays that can be used for macrophages, including:

1. Boyden Chamber Assay: This is a classic assay that involves placing macrophages in the upper chamber of a device separated by a porous membrane from a lower chamber containing a chemoattractant, such as a chemokine or growth factor. The macrophages migrate through the membrane towards the chemoattractant, and the number of cells that have migrated can be quantified.
2. Wound Healing Assay: In this assay, a "wound" is created in a confluent layer of macrophages, and the migration of the cells into the wound area is monitored over time. This assay can be used to study the effects of various factors on macrophage migration.
3. Transwell Assay: Similar to the Boyden Chamber assay, this assay involves placing macrophages in the upper chamber of a device separated by a porous membrane from a lower chamber containing a chemoattractant. However, in this case, the cells are allowed to migrate for a longer period of time, and the membrane is coated with a matrix protein, such as collagen or fibronectin, to better mimic the extracellular environment.
4. Scratch Assay: This assay involves creating a "scratch" in a confluent layer of macrophages and monitoring the migration of the cells into the scratch area over time. This assay can be used to study the effects of various factors on macrophage migration.

Overall, cell migration assays for macrophages are important tools for understanding the mechanisms that control macrophage movement and migration, which can have implications for a wide range of physiological and pathophysiological processes.

Cell division is the process by which a single eukaryotic cell (a cell with a true nucleus) divides into two identical daughter cells. This complex process involves several stages, including replication of DNA, separation of chromosomes, and division of the cytoplasm. There are two main types of cell division: mitosis and meiosis.

Mitosis is the type of cell division that results in two genetically identical daughter cells. It is a fundamental process for growth, development, and tissue repair in multicellular organisms. The stages of mitosis include prophase, prometaphase, metaphase, anaphase, and telophase, followed by cytokinesis, which divides the cytoplasm.

Meiosis, on the other hand, is a type of cell division that occurs in the gonads (ovaries and testes) during the production of gametes (sex cells). Meiosis results in four genetically unique daughter cells, each with half the number of chromosomes as the parent cell. This process is essential for sexual reproduction and genetic diversity. The stages of meiosis include meiosis I and meiosis II, which are further divided into prophase, prometaphase, metaphase, anaphase, and telophase.

In summary, cell division is the process by which a single cell divides into two daughter cells, either through mitosis or meiosis. This process is critical for growth, development, tissue repair, and sexual reproduction in multicellular organisms.

Cellular immunity, also known as cell-mediated immunity, is a type of immune response that involves the activation of immune cells, such as T lymphocytes (T cells), to protect the body against infected or damaged cells. This form of immunity is important for fighting off infections caused by viruses and intracellular bacteria, as well as for recognizing and destroying cancer cells.

Cellular immunity involves a complex series of interactions between various immune cells and molecules. When a pathogen infects a cell, the infected cell displays pieces of the pathogen on its surface in a process called antigen presentation. This attracts T cells, which recognize the antigens and become activated. Activated T cells then release cytokines, chemicals that help coordinate the immune response, and can directly attack and kill infected cells or help activate other immune cells to do so.

Cellular immunity is an important component of the adaptive immune system, which is able to learn and remember specific pathogens in order to mount a faster and more effective response upon subsequent exposure. This form of immunity is also critical for the rejection of transplanted organs, as the immune system recognizes the transplanted tissue as foreign and attacks it.

Siglec-1, also known as Sialic Acid Binding Ig-like Lectin 1, is a type of protein that belongs to the Siglec family. These proteins are found on the surface of certain immune cells, such as macrophages and dendritic cells, and they play a role in recognizing and binding to sialic acid molecules on other cells.

Siglec-1 is primarily expressed on the surface of monocytes and macrophages, and it has been shown to bind to sialic acids on the surface of various viruses, bacteria, and parasites. This binding can help to initiate an immune response against these pathogens. Siglec-1 has also been implicated in the development of certain inflammatory and autoimmune diseases, as well as in the progression of cancer.

In medical terms, Siglec-1 is often referred to by its molecular name, CD169 or Sialoadhesin, and it can be detected using various laboratory techniques such as flow cytometry, immunohistochemistry, and Western blotting.

Myeloid Differentiation Factor 88 (MYD88) is a signaling adaptor protein that plays a crucial role in the innate immune response. It is involved in the signal transduction pathways of several Toll-like receptors (TLRs), which are pattern recognition receptors that recognize pathogen-associated molecular patterns (PAMPs) and damage-associated molecular patterns (DAMPs).

Upon activation of TLRs, MYD88 is recruited to the receptor complex where it interacts with IL-1 receptor-associated kinase 4 (IRAK4) and activates IRAK1. This leads to the activation of downstream signaling pathways, including the mitogen-activated protein kinases (MAPKs) and nuclear factor kappa B (NF-κB), resulting in the production of proinflammatory cytokines and type I interferons.

MYD88 is widely expressed in various cell types, including hematopoietic cells, endothelial cells, and fibroblasts. Mutations in MYD88 have been associated with several human diseases, such as lymphomas, leukemias, and autoimmune disorders.

Chemotaxis is a term used in biology and medicine to describe the movement of an organism or cell towards or away from a chemical stimulus. This process plays a crucial role in various biological phenomena, including immune responses, wound healing, and the development and progression of diseases such as cancer.

In chemotaxis, cells can detect and respond to changes in the concentration of specific chemicals, known as chemoattractants or chemorepellents, in their environment. These chemicals bind to receptors on the cell surface, triggering a series of intracellular signaling events that ultimately lead to changes in the cytoskeleton and directed movement of the cell towards or away from the chemical gradient.

For example, during an immune response, white blood cells called neutrophils use chemotaxis to migrate towards sites of infection or inflammation, where they can attack and destroy invading pathogens. Similarly, cancer cells can use chemotaxis to migrate towards blood vessels and metastasize to other parts of the body.

Understanding chemotaxis is important for developing new therapies and treatments for a variety of diseases, including cancer, infectious diseases, and inflammatory disorders.

Cholesteryl esters are formed when cholesterol, a type of lipid (fat) that is important for the normal functioning of the body, becomes combined with fatty acids through a process called esterification. This results in a compound that is more hydrophobic (water-repelling) than cholesterol itself, which allows it to be stored more efficiently in the body.

Cholesteryl esters are found naturally in foods such as animal fats and oils, and they are also produced by the liver and other cells in the body. They play an important role in the structure and function of cell membranes, and they are also precursors to the synthesis of steroid hormones, bile acids, and vitamin D.

However, high levels of cholesteryl esters in the blood can contribute to the development of atherosclerosis, a condition characterized by the buildup of plaque in the arteries, which can increase the risk of heart disease and stroke. Cholesteryl esters are typically measured as part of a lipid profile, along with other markers such as total cholesterol, HDL cholesterol, and triglycerides.

In a medical context, nitrites are typically referred to as organic compounds that contain a functional group with the formula R-N=O, where R represents an alkyl or aryl group. They are commonly used in medicine as vasodilators, which means they widen and relax blood vessels, improving blood flow and lowering blood pressure.

One example of a nitrite used medically is amyl nitrite, which was previously used to treat angina pectoris, a type of chest pain caused by reduced blood flow to the heart muscle. However, its use has largely been replaced by other medications due to safety concerns and the availability of more effective treatments.

It's worth noting that inorganic nitrites, such as sodium nitrite, are also used in medicine for various purposes, including as a preservative in food and as a medication to treat cyanide poisoning. However, these compounds have different chemical properties and uses than organic nitrites.

HIV-1 (Human Immunodeficiency Virus type 1) is a species of the retrovirus genus that causes acquired immunodeficiency syndrome (AIDS). It is primarily transmitted through sexual contact, exposure to infected blood or blood products, and from mother to child during pregnancy, childbirth, or breastfeeding. HIV-1 infects vital cells in the human immune system, such as CD4+ T cells, macrophages, and dendritic cells, leading to a decline in their numbers and weakening of the immune response over time. This results in the individual becoming susceptible to various opportunistic infections and cancers that ultimately cause death if left untreated. HIV-1 is the most prevalent form of HIV worldwide and has been identified as the causative agent of the global AIDS pandemic.

Antigens are substances (usually proteins) on the surface of cells, viruses, fungi, or bacteria that can be recognized by the immune system and provoke an immune response. In the context of differentiation, antigens refer to specific markers that identify the developmental stage or lineage of a cell.

Differentiation antigens are proteins or carbohydrates expressed on the surface of cells during various stages of differentiation, which can be used to distinguish between cells at different maturation stages or of different cell types. These antigens play an essential role in the immune system's ability to recognize and respond to abnormal or infected cells while sparing healthy cells.

Examples of differentiation antigens include:

1. CD (cluster of differentiation) molecules: A group of membrane proteins used to identify and define various cell types, such as T cells, B cells, natural killer cells, monocytes, and granulocytes.
2. Lineage-specific antigens: Antigens that are specific to certain cell lineages, such as CD3 for T cells or CD19 for B cells.
3. Maturation markers: Antigens that indicate the maturation stage of a cell, like CD34 and CD38 on hematopoietic stem cells.

Understanding differentiation antigens is crucial in immunology, cancer research, transplantation medicine, and vaccine development.

Anti-inflammatory agents are a class of drugs or substances that reduce inflammation in the body. They work by inhibiting the production of inflammatory mediators, such as prostaglandins and leukotrienes, which are released during an immune response and contribute to symptoms like pain, swelling, redness, and warmth.

There are two main types of anti-inflammatory agents: steroidal and nonsteroidal. Steroidal anti-inflammatory drugs (SAIDs) include corticosteroids, which mimic the effects of hormones produced by the adrenal gland. Nonsteroidal anti-inflammatory drugs (NSAIDs) are a larger group that includes both prescription and over-the-counter medications, such as aspirin, ibuprofen, naproxen, and celecoxib.

While both types of anti-inflammatory agents can be effective in reducing inflammation and relieving symptoms, they differ in their mechanisms of action, side effects, and potential risks. Long-term use of NSAIDs, for example, can increase the risk of gastrointestinal bleeding, kidney damage, and cardiovascular events. Corticosteroids can have significant side effects as well, particularly with long-term use, including weight gain, mood changes, and increased susceptibility to infections.

It's important to use anti-inflammatory agents only as directed by a healthcare provider, and to be aware of potential risks and interactions with other medications or health conditions.

Interleukin-4 (IL-4) is a type of cytokine, which is a cell signaling molecule that mediates communication between cells in the immune system. Specifically, IL-4 is produced by activated T cells and mast cells, among other cells, and plays an important role in the differentiation and activation of immune cells called Th2 cells.

Th2 cells are involved in the immune response to parasites, as well as in allergic reactions. IL-4 also promotes the growth and survival of B cells, which produce antibodies, and helps to regulate the production of certain types of antibodies. In addition, IL-4 has anti-inflammatory effects and can help to downregulate the immune response in some contexts.

Defects in IL-4 signaling have been implicated in a number of diseases, including asthma, allergies, and certain types of cancer.

Tetradecanoylphorbol acetate (TPA) is defined as a pharmacological agent that is a derivative of the phorbol ester family. It is a potent tumor promoter and activator of protein kinase C (PKC), a group of enzymes that play a role in various cellular processes such as signal transduction, proliferation, and differentiation. TPA has been widely used in research to study PKC-mediated signaling pathways and its role in cancer development and progression. It is also used in topical treatments for skin conditions such as psoriasis.

Scavenger receptors, class B (SR-B) are a type of scavenger receptors that play a crucial role in the cellular uptake and metabolism of lipids, particularly modified low-density lipoproteins (LDL), high-density lipoproteins (HDL), and other lipid-soluble molecules. They are membrane-bound glycoproteins that contain an extracellular domain with a characteristic structure, including cysteine-rich repeats and transmembrane domains.

The best-characterized member of this class is SR-B1 (also known as CD36b, SCARB1), which is widely expressed in various tissues, such as the liver, steroidogenic organs, macrophages, and endothelial cells. SR-B1 selectively binds to HDL and facilitates the transfer of cholesteryl esters from HDL particles into cells while allowing HDL to maintain its structural integrity and continue its function in reverse cholesterol transport.

SR-B1 has also been implicated in the uptake and degradation of oxidized LDL, contributing to the development of atherosclerosis. Additionally, SR-B1 is involved in several other cellular processes, including innate immunity, inflammation, and angiogenesis.

Other members of class B scavenger receptors include SR-BI, SR-B2 (also known as CLA-1 or LIMPII), SR-B3 (also known as CD36c or SCARB2), and SR-B4 (also known as CXorf24). These receptors have distinct expression patterns and functions but share structural similarities with SR-BI.

In summary, scavenger receptors, class B, are a group of membrane-bound glycoproteins that facilitate the cellular uptake and metabolism of lipids, particularly modified LDL and HDL particles. They play essential roles in maintaining lipid homeostasis and have implications in various pathological conditions, such as atherosclerosis and inflammation.

Lymphocyte activation is the process by which B-cells and T-cells (types of lymphocytes) become activated to perform effector functions in an immune response. This process involves the recognition of specific antigens presented on the surface of antigen-presenting cells, such as dendritic cells or macrophages.

The activation of B-cells leads to their differentiation into plasma cells that produce antibodies, while the activation of T-cells results in the production of cytotoxic T-cells (CD8+ T-cells) that can directly kill infected cells or helper T-cells (CD4+ T-cells) that assist other immune cells.

Lymphocyte activation involves a series of intracellular signaling events, including the binding of co-stimulatory molecules and the release of cytokines, which ultimately result in the expression of genes involved in cell proliferation, differentiation, and effector functions. The activation process is tightly regulated to prevent excessive or inappropriate immune responses that can lead to autoimmunity or chronic inflammation.

Cell migration inhibition refers to the process or agents that restrict the movement of cells, particularly in the context of cancer metastasis. Cell migration is a critical biological process involved in various physiological and pathological conditions, including embryonic development, wound healing, and tumor cell dissemination. Inhibiting cell migration can help prevent the spread of cancer to distant organs, thereby improving treatment outcomes and patient survival rates.

Various factors and mechanisms contribute to cell migration inhibition, such as:

1. Modulation of signaling pathways: Cell migration is regulated by complex intracellular signaling networks that control cytoskeletal rearrangements, adhesion molecules, and other components required for cell motility. Inhibiting specific signaling proteins or pathways can suppress cell migration.
2. Extracellular matrix (ECM) modifications: The ECM provides structural support and biochemical cues that guide cell migration. Altering the composition or organization of the ECM can hinder cell movement.
3. Inhibition of adhesion molecules: Cell-cell and cell-matrix interactions are mediated by adhesion molecules, such as integrins and cadherins. Blocking these molecules can prevent cells from attaching to their surroundings and migrating.
4. Targeting cytoskeletal components: The cytoskeleton is responsible for the mechanical forces required for cell migration. Inhibiting cytoskeletal proteins, such as actin or tubulin, can impair cell motility.
5. Use of pharmacological agents: Several drugs and compounds have been identified to inhibit cell migration, either by targeting specific molecules or indirectly affecting the overall cellular environment. These agents include chemotherapeutic drugs, natural compounds, and small molecule inhibitors.

Understanding the mechanisms underlying cell migration inhibition can provide valuable insights into developing novel therapeutic strategies for cancer treatment and other diseases involving aberrant cell migration.

Cell separation is a process used to separate and isolate specific cell types from a heterogeneous mixture of cells. This can be accomplished through various physical or biological methods, depending on the characteristics of the cells of interest. Some common techniques for cell separation include:

1. Density gradient centrifugation: In this method, a sample containing a mixture of cells is layered onto a density gradient medium and then centrifuged. The cells are separated based on their size, density, and sedimentation rate, with denser cells settling closer to the bottom of the tube and less dense cells remaining near the top.

2. Magnetic-activated cell sorting (MACS): This technique uses magnetic beads coated with antibodies that bind to specific cell surface markers. The labeled cells are then passed through a column placed in a magnetic field, which retains the magnetically labeled cells while allowing unlabeled cells to flow through.

3. Fluorescence-activated cell sorting (FACS): In this method, cells are stained with fluorochrome-conjugated antibodies that recognize specific cell surface or intracellular markers. The stained cells are then passed through a laser beam, which excites the fluorophores and allows for the detection and sorting of individual cells based on their fluorescence profile.

4. Filtration: This simple method relies on the physical size differences between cells to separate them. Cells can be passed through filters with pore sizes that allow smaller cells to pass through while retaining larger cells.

5. Enzymatic digestion: In some cases, cells can be separated by enzymatically dissociating tissues into single-cell suspensions and then using various separation techniques to isolate specific cell types.

These methods are widely used in research and clinical settings for applications such as isolating immune cells, stem cells, or tumor cells from biological samples.

"CBA" is an abbreviation for a specific strain of inbred mice that were developed at the Cancer Research Institute in London. The "Inbred CBA" mice are genetically identical individuals within the same strain, due to many generations of brother-sister matings. This results in a homozygous population, making them valuable tools for research because they reduce variability and increase reproducibility in experimental outcomes.

The CBA strain is known for its susceptibility to certain diseases, such as autoimmune disorders and cancer, which makes it a popular choice for researchers studying those conditions. Additionally, the CBA strain has been widely used in studies related to transplantation immunology, infectious diseases, and genetic research.

It's important to note that while "Inbred CBA" mice are a well-established and useful tool in biomedical research, they represent only one of many inbred strains available for scientific investigation. Each strain has its own unique characteristics and advantages, depending on the specific research question being asked.

Leukocytes, also known as white blood cells (WBCs), are a crucial component of the human immune system. They are responsible for protecting the body against infections and foreign substances. Leukocytes are produced in the bone marrow and circulate throughout the body in the bloodstream and lymphatic system.

There are several types of leukocytes, including:

1. Neutrophils - These are the most abundant type of leukocyte and are primarily responsible for fighting bacterial infections. They contain enzymes that can destroy bacteria.
2. Lymphocytes - These are responsible for producing antibodies and destroying virus-infected cells, as well as cancer cells. There are two main types of lymphocytes: B-lymphocytes and T-lymphocytes.
3. Monocytes - These are the largest type of leukocyte and help to break down and remove dead or damaged tissues, as well as microorganisms.
4. Eosinophils - These play a role in fighting parasitic infections and are also involved in allergic reactions and inflammation.
5. Basophils - These release histamine and other chemicals that cause inflammation in response to allergens or irritants.

An abnormal increase or decrease in the number of leukocytes can indicate an underlying medical condition, such as an infection, inflammation, or a blood disorder.

Leishmania is a genus of protozoan parasites that are the causative agents of Leishmaniasis, a group of diseases with various clinical manifestations. These parasites are transmitted to humans through the bite of infected female phlebotomine sandflies. The disease has a wide geographic distribution, mainly in tropical and subtropical regions, including parts of Asia, Africa, South America, and Southern Europe.

The Leishmania species have a complex life cycle that involves two main stages: the promastigote stage, which is found in the sandfly vector, and the amastigote stage, which infects mammalian hosts, including humans. The clinical manifestations of Leishmaniasis depend on the specific Leishmania species and the host's immune response to the infection.

The three main forms of Leishmaniasis are:

1. Cutaneous Leishmaniasis (CL): This form is characterized by skin lesions, such as ulcers or nodules, that can take several months to heal and may leave scars. CL is caused by various Leishmania species, including L. major, L. tropica, and L. aethiopica.

2. Visceral Leishmaniasis (VL): Also known as kala-azar, VL affects internal organs such as the spleen, liver, and bone marrow. Symptoms include fever, weight loss, anemia, and enlarged liver and spleen. VL is caused by L. donovani, L. infantum, and L. chagasi species.

3. Mucocutaneous Leishmaniasis (MCL): This form affects the mucous membranes of the nose, mouth, and throat, causing destruction of tissues and severe disfigurement. MCL is caused by L. braziliensis and L. guyanensis species.

Prevention and control measures for Leishmaniasis include vector control, early diagnosis and treatment, and protection against sandfly bites through the use of insect repellents and bed nets.

ICR (Institute of Cancer Research) is a strain of albino Swiss mice that are widely used in scientific research. They are an outbred strain, which means that they have been bred to maintain maximum genetic heterogeneity. However, it is also possible to find inbred strains of ICR mice, which are genetically identical individuals produced by many generations of brother-sister mating.

Inbred ICR mice are a specific type of ICR mouse that has been inbred for at least 20 generations. This means that they have a high degree of genetic uniformity and are essentially genetically identical to one another. Inbred strains of mice are often used in research because their genetic consistency makes them more reliable models for studying biological phenomena and testing new therapies or treatments.

It is important to note that while inbred ICR mice may be useful for certain types of research, they do not necessarily represent the genetic diversity found in human populations. Therefore, it is important to consider the limitations of using any animal model when interpreting research findings and applying them to human health.

Chemotactic factors are substances that attract or repel cells, particularly immune cells, by stimulating directional movement in response to a chemical gradient. These factors play a crucial role in the body's immune response and inflammation process. They include:

1. Chemokines: A family of small signaling proteins that direct the migration of immune cells to sites of infection or tissue damage.
2. Cytokines: A broad category of signaling molecules that mediate and regulate immunity, inflammation, and hematopoiesis. Some cytokines can also act as chemotactic factors.
3. Complement components: Cleavage products of the complement system can attract immune cells to the site of infection or tissue injury.
4. Growth factors: Certain growth factors, like colony-stimulating factors (CSFs), can stimulate the migration and proliferation of specific cell types.
5. Lipid mediators: Products derived from arachidonic acid metabolism, such as leukotrienes and prostaglandins, can also act as chemotactic factors.
6. Formyl peptides: Bacterial-derived formylated peptides can attract and activate neutrophils during an infection.
7. Extracellular matrix (ECM) components: Fragments of ECM proteins, like collagen and fibronectin, can serve as chemotactic factors for immune cells.

These factors help orchestrate the immune response by guiding the movement of immune cells to specific locations in the body where they are needed.

Enzyme activation refers to the process by which an enzyme becomes biologically active and capable of carrying out its specific chemical or biological reaction. This is often achieved through various post-translational modifications, such as proteolytic cleavage, phosphorylation, or addition of cofactors or prosthetic groups to the enzyme molecule. These modifications can change the conformation or structure of the enzyme, exposing or creating a binding site for the substrate and allowing the enzymatic reaction to occur.

For example, in the case of proteolytic cleavage, an inactive precursor enzyme, known as a zymogen, is cleaved into its active form by a specific protease. This is seen in enzymes such as trypsin and chymotrypsin, which are initially produced in the pancreas as inactive precursors called trypsinogen and chymotrypsinogen, respectively. Once they reach the small intestine, they are activated by enteropeptidase, a protease that cleaves a specific peptide bond, releasing the active enzyme.

Phosphorylation is another common mechanism of enzyme activation, where a phosphate group is added to a specific serine, threonine, or tyrosine residue on the enzyme by a protein kinase. This modification can alter the conformation of the enzyme and create a binding site for the substrate, allowing the enzymatic reaction to occur.

Enzyme activation is a crucial process in many biological pathways, as it allows for precise control over when and where specific reactions take place. It also provides a mechanism for regulating enzyme activity in response to various signals and stimuli, such as hormones, neurotransmitters, or changes in the intracellular environment.

Propionibacterium acnes is a gram-positive, rod-shaped bacterium that naturally colonizes the skin, predominantly in areas with a high density of sebaceous glands such as the face, back, and chest. It is part of the normal skin flora but can contribute to the development of acne vulgaris when it proliferates excessively and clogs the pilosebaceous units (hair follicles).

The bacterium metabolizes sebum, producing propionic acid and other short-chain fatty acids as byproducts. In acne, these byproducts can cause an inflammatory response in the skin, leading to the formation of papules, pustules, and nodules. Propionibacterium acnes has also been implicated in various other skin conditions and occasionally in opportunistic infections in other parts of the body, particularly in immunocompromised individuals or following surgical procedures.

Silicon dioxide is not a medical term, but a chemical compound with the formula SiO2. It's commonly known as quartz or sand and is not something that would typically have a medical definition. However, in some cases, silicon dioxide can be used in pharmaceutical preparations as an excipient (an inactive substance that serves as a vehicle or medium for a drug) or as a food additive, often as an anti-caking agent.

In these contexts, it's important to note that silicon dioxide is considered generally recognized as safe (GRAS) by the U.S. Food and Drug Administration (FDA). However, exposure to very high levels of respirable silica dust, such as in certain industrial settings, can increase the risk of lung disease, including silicosis.

LDL receptors (Low-Density Lipoprotein Receptors) are cell surface receptors that play a crucial role in the regulation of cholesterol homeostasis within the body. They are responsible for recognizing and binding to LDL particles, also known as "bad cholesterol," which are then internalized by the cell through endocytosis.

Once inside the cell, the LDL particles are broken down, releasing their cholesterol content, which can be used for various cellular processes such as membrane synthesis and hormone production. The LDL receptors themselves are recycled back to the cell surface, allowing for continued uptake of LDL particles.

Mutations in the LDL receptor gene can lead to a condition called familial hypercholesterolemia, which is characterized by high levels of LDL cholesterol in the blood and an increased risk of premature cardiovascular disease.

The liver is a large, solid organ located in the upper right portion of the abdomen, beneath the diaphragm and above the stomach. It plays a vital role in several bodily functions, including:

1. Metabolism: The liver helps to metabolize carbohydrates, fats, and proteins from the food we eat into energy and nutrients that our bodies can use.
2. Detoxification: The liver detoxifies harmful substances in the body by breaking them down into less toxic forms or excreting them through bile.
3. Synthesis: The liver synthesizes important proteins, such as albumin and clotting factors, that are necessary for proper bodily function.
4. Storage: The liver stores glucose, vitamins, and minerals that can be released when the body needs them.
5. Bile production: The liver produces bile, a digestive juice that helps to break down fats in the small intestine.
6. Immune function: The liver plays a role in the immune system by filtering out bacteria and other harmful substances from the blood.

Overall, the liver is an essential organ that plays a critical role in maintaining overall health and well-being.

Chemotaxis, Leukocyte is the movement of leukocytes (white blood cells) towards a higher concentration of a particular chemical substance, known as a chemotactic factor. This process plays a crucial role in the immune system's response to infection and injury.

When there is an infection or tissue damage, certain cells release chemotactic factors, which are small molecules or proteins that can attract leukocytes to the site of inflammation. Leukocytes have receptors on their surface that can detect these chemotactic factors and move towards them through a process called chemotaxis.

Once they reach the site of inflammation, leukocytes can help eliminate pathogens or damaged cells by phagocytosis (engulfing and destroying) or releasing toxic substances that kill the invading microorganisms. Chemotaxis is an essential part of the immune system's defense mechanisms and helps to maintain tissue homeostasis and prevent the spread of infection.

Cell communication, also known as cell signaling, is the process by which cells exchange and transmit signals between each other and their environment. This complex system allows cells to coordinate their functions and maintain tissue homeostasis. Cell communication can occur through various mechanisms including:

1. Autocrine signaling: When a cell releases a signal that binds to receptors on the same cell, leading to changes in its behavior or function.
2. Paracrine signaling: When a cell releases a signal that binds to receptors on nearby cells, influencing their behavior or function.
3. Endocrine signaling: When a cell releases a hormone into the bloodstream, which then travels to distant target cells and binds to specific receptors, triggering a response.
4. Synaptic signaling: In neurons, communication occurs through the release of neurotransmitters that cross the synapse and bind to receptors on the postsynaptic cell, transmitting electrical or chemical signals.
5. Contact-dependent signaling: When cells physically interact with each other, allowing for the direct exchange of signals and information.

Cell communication is essential for various physiological processes such as growth, development, differentiation, metabolism, immune response, and tissue repair. Dysregulation in cell communication can contribute to diseases, including cancer, diabetes, and neurological disorders.

Caspase-1 is a type of protease enzyme that plays a crucial role in the inflammatory response and programmed cell death, also known as apoptosis. It is produced as an inactive precursor protein, which is then cleaved into its active form by other proteases or through self-cleavage.

Once activated, caspase-1 helps to process and activate several pro-inflammatory cytokines, such as interleukin (IL)-1β and IL-18, which are involved in the recruitment of immune cells to sites of infection or tissue damage. Caspase-1 also contributes to programmed cell death by cleaving and activating other caspases, leading to the controlled destruction of the cell.

Dysregulation of caspase-1 has been implicated in various inflammatory diseases, such as autoimmune disorders and neurodegenerative conditions. Therefore, understanding the mechanisms that regulate caspase-1 activity is an important area of research for developing new therapeutic strategies to treat these diseases.

Endotoxins are toxic substances that are associated with the cell walls of certain types of bacteria. They are released when the bacterial cells die or divide, and can cause a variety of harmful effects in humans and animals. Endotoxins are made up of lipopolysaccharides (LPS), which are complex molecules consisting of a lipid and a polysaccharide component.

Endotoxins are particularly associated with gram-negative bacteria, which have a distinctive cell wall structure that includes an outer membrane containing LPS. These toxins can cause fever, inflammation, and other symptoms when they enter the bloodstream or other tissues of the body. They are also known to play a role in the development of sepsis, a potentially life-threatening condition characterized by a severe immune response to infection.

Endotoxins are resistant to heat, acid, and many disinfectants, making them difficult to eliminate from contaminated environments. They can also be found in a variety of settings, including hospitals, industrial facilities, and agricultural operations, where they can pose a risk to human health.

Cyclooxygenase-2 (COX-2) is an enzyme involved in the synthesis of prostaglandins, which are hormone-like substances that play a role in inflammation, pain, and fever. COX-2 is primarily expressed in response to stimuli such as cytokines and growth factors, and its expression is associated with the development of inflammation.

COX-2 inhibitors are a class of nonsteroidal anti-inflammatory drugs (NSAIDs) that selectively block the activity of COX-2, reducing the production of prostaglandins and providing analgesic, anti-inflammatory, and antipyretic effects. These medications are often used to treat pain and inflammation associated with conditions such as arthritis, menstrual cramps, and headaches.

It's important to note that while COX-2 inhibitors can be effective in managing pain and inflammation, they may also increase the risk of cardiovascular events such as heart attack and stroke, particularly when used at high doses or for extended periods. Therefore, it's essential to use these medications under the guidance of a healthcare provider and to follow their instructions carefully.

Complement receptors are proteins found on the surface of various cells in the human body, including immune cells and some non-immune cells. They play a crucial role in the complement system, which is a part of the innate immune response that helps to eliminate pathogens and damaged cells from the body. Complement receptors bind to complement proteins or fragments that are generated during the activation of the complement system. This binding triggers various intracellular signaling events that can lead to diverse cellular responses, such as phagocytosis, inflammation, and immune regulation.

There are several types of complement receptors, including:

1. CR1 (CD35): A receptor found on erythrocytes, B cells, neutrophils, monocytes, macrophages, and glomerular podocytes. It functions in the clearance of immune complexes and regulates complement activation.
2. CR2 (CD21): Expressed mainly on B cells and follicular dendritic cells. It facilitates antigen presentation, B-cell activation, and immune regulation.
3. CR3 (CD11b/CD18, Mac-1): Present on neutrophils, monocytes, macrophages, and some T cells. It mediates cell adhesion, phagocytosis, and intracellular signaling.
4. CR4 (CD11c/CD18, p150,95): Expressed on neutrophils, monocytes, macrophages, and dendritic cells. It is involved in cell adhesion, phagocytosis, and intracellular signaling.
5. C5aR (CD88): Found on various immune cells, including neutrophils, monocytes, macrophages, mast cells, eosinophils, and dendritic cells. It binds to the complement protein C5a and mediates chemotaxis, degranulation, and inflammation.
6. C5L2 (GPR77): Present on various cell types, including immune cells. Its function is not well understood but may involve regulating C5a-mediated responses or acting as a receptor for other ligands.

These receptors play crucial roles in the immune response and inflammation by mediating various functions such as chemotaxis, phagocytosis, cell adhesion, and intracellular signaling. Dysregulation of these receptors has been implicated in several diseases, including autoimmune disorders, infections, and cancer.

Acid phosphatase is a type of enzyme that is found in various tissues and organs throughout the body, including the prostate gland, red blood cells, bone, liver, spleen, and kidneys. This enzyme plays a role in several biological processes, such as bone metabolism and the breakdown of molecules like nucleotides and proteins.

Acid phosphatase is classified based on its optimum pH level for activity. Acid phosphatases have an optimal activity at acidic pH levels (below 7.0), while alkaline phosphatases have an optimal activity at basic or alkaline pH levels (above 7.0).

In clinical settings, measuring the level of acid phosphatase in the blood can be useful as a tumor marker for prostate cancer. Elevated acid phosphatase levels may indicate the presence of metastatic prostate cancer or disease progression. However, it is important to note that acid phosphatase is not specific to prostate cancer and can also be elevated in other conditions, such as bone diseases, liver disorders, and some benign conditions. Therefore, acid phosphatase should be interpreted in conjunction with other diagnostic tests and clinical findings for a more accurate diagnosis.

Lipoprotein receptors are specialized proteins found on the surface of cells that play a crucial role in the metabolism of lipoproteins, which are complex particles composed of lipids and proteins. These receptors bind to specific lipoproteins in the bloodstream, facilitating their uptake into the cell for further processing.

There are several types of lipoprotein receptors, including:

1. LDL (Low-Density Lipoprotein) Receptor: This receptor is responsible for recognizing and internalizing LDL particles, which are rich in cholesterol. Once inside the cell, LDL particles release their cholesterol, which can then be used for various cellular functions or stored for later use. Defects in the LDL receptor can lead to elevated levels of LDL cholesterol in the blood and an increased risk of developing cardiovascular disease.
2. HDL (High-Density Lipoprotein) Receptor: This receptor is involved in the clearance of HDL particles from the bloodstream. HDL particles are responsible for transporting excess cholesterol from peripheral tissues to the liver, where it can be processed and eliminated from the body.
3. VLDL (Very Low-Density Lipoprotein) Receptor: This receptor recognizes and internalizes VLDL particles, which are produced by the liver and carry triglycerides and cholesterol to peripheral tissues. VLDL particles are subsequently converted into LDL particles in the bloodstream.
4. LRP (Low-Density Lipoprotein Receptor-Related Protein) Family: This family of receptors includes several members, such as LRP1 and LRP2, that play roles in various cellular processes, including lipid metabolism, protein trafficking, and cell signaling. They can bind to a variety of ligands, including lipoproteins, proteases, and extracellular matrix components.

In summary, lipoprotein receptors are essential for maintaining proper lipid metabolism and homeostasis by facilitating the uptake, processing, and elimination of lipoproteins in the body.

CCR2 (C-C chemokine receptor type 2) is a type of protein found on the surface of certain immune cells, including monocytes and memory T cells. It is a type of G protein-coupled receptor that binds to specific chemokines, which are small signaling proteins that help regulate the movement of immune cells throughout the body.

CCR2 plays an important role in the immune response by mediating the migration of monocytes and other immune cells to sites of inflammation or injury. When a chemokine binds to CCR2, it triggers a series of intracellular signaling events that cause the cell to move towards the source of the chemokine.

In addition to its role in the immune response, CCR2 has been implicated in various disease processes, including atherosclerosis, rheumatoid arthritis, and cancer metastasis. In these contexts, CCR2 antagonists have been explored as potential therapeutic agents to block the recruitment of immune cells and reduce inflammation or tumor growth.

Listeriosis is an infection caused by the bacterium Listeria monocytogenes. It primarily affects older adults, individuals with weakened immune systems, pregnant women, and newborns. The bacteria can be found in contaminated food, water, or soil. Symptoms of listeriosis may include fever, muscle aches, headache, stiff neck, confusion, loss of balance, and convulsions. In severe cases, it can lead to meningitis (inflammation of the membranes surrounding the brain and spinal cord) or bacteremia (bacterial infection in the bloodstream). Pregnant women may experience only mild flu-like symptoms, but listeriosis can lead to miscarriage, stillbirth, premature delivery, or serious illness in newborns.

It's important to note that listeriosis is a foodborne illness, and proper food handling, cooking, and storage practices can help prevent infection. High-risk individuals should avoid consuming unpasteurized dairy products, raw or undercooked meat, poultry, and seafood, as well as soft cheeses made from unpasteurized milk.

Colony-stimulating factors (CSFs) are a group of growth factors that stimulate the production of blood cells in the bone marrow. They include granulocyte colony-stimulating factor (G-CSF), granulocyte-macrophage colony-stimulating factor (GM-CSF), and macrophage colony-stimulating factor (M-CSF). These factors play an important role in the regulation of hematopoiesis, which is the process of producing different types of blood cells.

G-CSF stimulates the production of neutrophils, a type of white blood cell that helps fight against bacterial and fungal infections. GM-CSF stimulates the production of both neutrophils and monocytes/macrophages, which are important in the immune response to infection and tissue injury. M-CSF stimulates the production and activation of macrophages, which play a role in the immune response, wound healing, and the regulation of hematopoiesis.

Colony-stimulating factors are used clinically to stimulate the production of white blood cells in patients undergoing chemotherapy or radiation therapy, which can suppress bone marrow function and lead to low white blood cell counts. They are also used to mobilize stem cells from the bone marrow into the peripheral blood for collection and transplantation.

A "mutant strain of mice" in a medical context refers to genetically engineered mice that have specific genetic mutations introduced into their DNA. These mutations can be designed to mimic certain human diseases or conditions, allowing researchers to study the underlying biological mechanisms and test potential therapies in a controlled laboratory setting.

Mutant strains of mice are created through various techniques, including embryonic stem cell manipulation, gene editing technologies such as CRISPR-Cas9, and radiation-induced mutagenesis. These methods allow scientists to introduce specific genetic changes into the mouse genome, resulting in mice that exhibit altered physiological or behavioral traits.

These strains of mice are widely used in biomedical research because their short lifespan, small size, and high reproductive rate make them an ideal model organism for studying human diseases. Additionally, the mouse genome has been well-characterized, and many genetic tools and resources are available to researchers working with these animals.

Examples of mutant strains of mice include those that carry mutations in genes associated with cancer, neurodegenerative disorders, metabolic diseases, and immunological conditions. These mice provide valuable insights into the pathophysiology of human diseases and help advance our understanding of potential therapeutic interventions.

In a medical context, "latex" refers to the natural rubber milk-like substance that is tapped from the incisions made in the bark of the rubber tree (Hevea brasiliensis). This sap is then processed to create various products such as gloves, catheters, and balloons. It's important to note that some people may have a latex allergy, which can cause mild to severe reactions when they come into contact with latex products.

Reactive Oxygen Species (ROS) are highly reactive molecules containing oxygen, including peroxides, superoxide, hydroxyl radical, and singlet oxygen. They are naturally produced as byproducts of normal cellular metabolism in the mitochondria, and can also be generated by external sources such as ionizing radiation, tobacco smoke, and air pollutants. At low or moderate concentrations, ROS play important roles in cell signaling and homeostasis, but at high concentrations, they can cause significant damage to cell structures, including lipids, proteins, and DNA, leading to oxidative stress and potential cell death.

Respiratory burst is a term used in the field of biology, particularly in the context of immunology and cellular processes. It does not have a direct application to clinical medicine, but it is important for understanding certain physiological and pathophysiological mechanisms. Here's a definition of respiratory burst:

Respiratory burst is a rapid increase in oxygen consumption by phagocytic cells (like neutrophils, monocytes, and macrophages) following their activation in response to various stimuli, such as pathogens or inflammatory molecules. This process is part of the innate immune response and serves to eliminate invading microorganisms.

The respiratory burst involves the activation of NADPH oxidase, an enzyme complex present in the membrane of phagosomes (the compartment where pathogens are engulfed). Upon activation, NADPH oxidase catalyzes the reduction of oxygen to superoxide radicals, which then dismutate to form hydrogen peroxide. These reactive oxygen species (ROS) can directly kill or damage microorganisms and also serve as signaling molecules for other immune cells.

While respiratory burst is a crucial part of the immune response, excessive or dysregulated ROS production can contribute to tissue damage and chronic inflammation, which have implications in various pathological conditions, such as atherosclerosis, neurodegenerative diseases, and cancer.

Enzyme inhibitors are substances that bind to an enzyme and decrease its activity, preventing it from catalyzing a chemical reaction in the body. They can work by several mechanisms, including blocking the active site where the substrate binds, or binding to another site on the enzyme to change its shape and prevent substrate binding. Enzyme inhibitors are often used as drugs to treat various medical conditions, such as high blood pressure, abnormal heart rhythms, and bacterial infections. They can also be found naturally in some foods and plants, and can be used in research to understand enzyme function and regulation.

Carrier proteins, also known as transport proteins, are a type of protein that facilitates the movement of molecules across cell membranes. They are responsible for the selective and active transport of ions, sugars, amino acids, and other molecules from one side of the membrane to the other, against their concentration gradient. This process requires energy, usually in the form of ATP (adenosine triphosphate).

Carrier proteins have a specific binding site for the molecule they transport, and undergo conformational changes upon binding, which allows them to move the molecule across the membrane. Once the molecule has been transported, the carrier protein returns to its original conformation, ready to bind and transport another molecule.

Carrier proteins play a crucial role in maintaining the balance of ions and other molecules inside and outside of cells, and are essential for many physiological processes, including nerve impulse transmission, muscle contraction, and nutrient uptake.

Leishmania mexicana is a species of protozoan parasite that causes cutaneous leishmaniasis, a skin infection, in humans and other mammals. It is transmitted to its hosts through the bite of infected female sandflies, primarily of the genus Lutzomyia. The parasites multiply within the skin lesions of the host, leading to symptoms such as ulcers, scarring, and disfigurement. The severity and duration of the infection can vary widely, and in some cases, the infection may heal on its own without treatment. However, in other cases, the infection can become chronic and lead to significant morbidity.

Leishmania mexicana is found primarily in Mexico and Central America, although it has also been reported in other parts of the world. It is one of several species of Leishmania that can cause cutaneous leishmaniasis, and diagnosis typically involves identifying the parasite through microscopic examination of tissue samples or through molecular testing. Treatment options for cutaneous leishmaniasis caused by L. mexicana include systemic medications such as antimony compounds, miltefosine, and amphotericin B, as well as local treatments such as heat therapy and cryotherapy.

Liposomes are artificially prepared, small, spherical vesicles composed of one or more lipid bilayers that enclose an aqueous compartment. They can encapsulate both hydrophilic and hydrophobic drugs, making them useful for drug delivery applications in the medical field. The lipid bilayer structure of liposomes is similar to that of biological membranes, which allows them to merge with and deliver their contents into cells. This property makes liposomes a valuable tool in delivering drugs directly to targeted sites within the body, improving drug efficacy while minimizing side effects.

Intramolecular oxidoreductases are a specific class of enzymes that catalyze the transfer of electrons within a single molecule, hence the term "intramolecular." These enzymes are involved in oxidoreduction reactions, where one part of the molecule is oxidized (loses electrons) and another part is reduced (gains electrons). This process allows for the rearrangement or modification of functional groups within the molecule.

The term "oxidoreductase" refers to enzymes that catalyze oxidation-reduction reactions, which are also known as redox reactions. These enzymes play a crucial role in various biological processes, including energy metabolism, detoxification, and biosynthesis.

It's important to note that intramolecular oxidoreductases should not be confused with intermolecular oxidoreductases, which catalyze redox reactions between two separate molecules.

p38 Mitogen-Activated Protein Kinases (p38 MAPKs) are a family of conserved serine-threonine protein kinases that play crucial roles in various cellular processes, including inflammation, immune response, differentiation, apoptosis, and stress responses. They are activated by diverse stimuli such as cytokines, ultraviolet radiation, heat shock, osmotic stress, and lipopolysaccharides (LPS).

Once activated, p38 MAPKs phosphorylate and regulate several downstream targets, including transcription factors and other protein kinases. This regulation leads to the expression of genes involved in inflammation, cell cycle arrest, and apoptosis. Dysregulation of p38 MAPK signaling has been implicated in various diseases, such as cancer, neurodegenerative disorders, and autoimmune diseases. Therefore, p38 MAPKs are considered promising targets for developing new therapeutic strategies to treat these conditions.

'Leishmania donovani' is a species of protozoan parasite that causes a severe form of visceral leishmaniasis, also known as kala-azar. This disease primarily affects the spleen, liver, and bone marrow, leading to symptoms such as fever, weight loss, anemia, and enlargement of the spleen and liver. The parasite is transmitted to humans through the bite of infected female sandflies. It's worth noting that this organism can also affect dogs and other animals, causing a disease known as canine leishmaniasis.

ATP-binding cassette (ABC) transporters are a family of membrane proteins that utilize the energy from ATP hydrolysis to transport various substrates across extra- and intracellular membranes. These transporters play crucial roles in several biological processes, including detoxification, drug resistance, nutrient uptake, and regulation of cellular cholesterol homeostasis.

The structure of ABC transporters consists of two nucleotide-binding domains (NBDs) that bind and hydrolyze ATP, and two transmembrane domains (TMDs) that form the substrate-translocation pathway. The NBDs are typically located adjacent to each other in the cytoplasm, while the TMDs can be either integral membrane domains or separate structures associated with the membrane.

The human genome encodes 48 distinct ABC transporters, which are classified into seven subfamilies (ABCA-ABCG) based on their sequence similarity and domain organization. Some well-known examples of ABC transporters include P-glycoprotein (ABCB1), multidrug resistance protein 1 (ABCC1), and breast cancer resistance protein (ABCG2).

Dysregulation or mutations in ABC transporters have been implicated in various diseases, such as cystic fibrosis, neurological disorders, and cancer. In cancer, overexpression of certain ABC transporters can contribute to drug resistance by actively effluxing chemotherapeutic agents from cancer cells, making them less susceptible to treatment.

Interleukin-8 (IL-8) is a type of cytokine, which is a small signaling protein involved in immune response and inflammation. IL-8 is also known as neutrophil chemotactic factor or NCF because it attracts neutrophils, a type of white blood cell, to the site of infection or injury.

IL-8 is produced by various cells including macrophages, epithelial cells, and endothelial cells in response to bacterial or inflammatory stimuli. It acts by binding to specific receptors called CXCR1 and CXCR2 on the surface of neutrophils, which triggers a series of intracellular signaling events leading to neutrophil activation, migration, and degranulation.

IL-8 plays an important role in the recruitment of neutrophils to the site of infection or tissue damage, where they can phagocytose and destroy invading microorganisms. However, excessive or prolonged production of IL-8 has been implicated in various inflammatory diseases such as chronic obstructive pulmonary disease (COPD), rheumatoid arthritis, and cancer.

Enzyme induction is a process by which the activity or expression of an enzyme is increased in response to some stimulus, such as a drug, hormone, or other environmental factor. This can occur through several mechanisms, including increasing the transcription of the enzyme's gene, stabilizing the mRNA that encodes the enzyme, or increasing the translation of the mRNA into protein.

In some cases, enzyme induction can be a beneficial process, such as when it helps the body to metabolize and clear drugs more quickly. However, in other cases, enzyme induction can have negative consequences, such as when it leads to the increased metabolism of important endogenous compounds or the activation of harmful procarcinogens.

Enzyme induction is an important concept in pharmacology and toxicology, as it can affect the efficacy and safety of drugs and other xenobiotics. It is also relevant to the study of drug interactions, as the induction of one enzyme by a drug can lead to altered metabolism and effects of another drug that is metabolized by the same enzyme.

The Fluorescent Antibody Technique (FAT) is a type of immunofluorescence assay used in laboratory medicine and pathology for the detection and localization of specific antigens or antibodies in tissues, cells, or microorganisms. In this technique, a fluorescein-labeled antibody is used to selectively bind to the target antigen or antibody, forming an immune complex. When excited by light of a specific wavelength, the fluorescein label emits light at a longer wavelength, typically visualized as green fluorescence under a fluorescence microscope.

The FAT is widely used in diagnostic microbiology for the identification and characterization of various bacteria, viruses, fungi, and parasites. It has also been applied in the diagnosis of autoimmune diseases and certain cancers by detecting specific antibodies or antigens in patient samples. The main advantage of FAT is its high sensitivity and specificity, allowing for accurate detection and differentiation of various pathogens and disease markers. However, it requires specialized equipment and trained personnel to perform and interpret the results.

Mononuclear leukocytes are a type of white blood cells (leukocytes) that have a single, large nucleus. They include lymphocytes (B-cells, T-cells, and natural killer cells), monocytes, and dendritic cells. These cells play important roles in the body's immune system, including defending against infection and disease, and participating in immune responses and surveillance. Mononuclear leukocytes can be found in the bloodstream as well as in tissues throughout the body. They are involved in both innate and adaptive immunity, providing specific and nonspecific defense mechanisms to protect the body from harmful pathogens and other threats.

Phosphorylation is the process of adding a phosphate group (a molecule consisting of one phosphorus atom and four oxygen atoms) to a protein or other organic molecule, which is usually done by enzymes called kinases. This post-translational modification can change the function, localization, or activity of the target molecule, playing a crucial role in various cellular processes such as signal transduction, metabolism, and regulation of gene expression. Phosphorylation is reversible, and the removal of the phosphate group is facilitated by enzymes called phosphatases.

Hydrogen peroxide (H2O2) is a colorless, odorless, clear liquid with a slightly sweet taste, although drinking it is harmful and can cause poisoning. It is a weak oxidizing agent and is used as an antiseptic and a bleaching agent. In diluted form, it is used to disinfect wounds and kill bacteria and viruses on the skin; in higher concentrations, it can be used to bleach hair or remove stains from clothing. It is also used as a propellant in rocketry and in certain industrial processes. Chemically, hydrogen peroxide is composed of two hydrogen atoms and two oxygen atoms, and it is structurally similar to water (H2O), with an extra oxygen atom. This gives it its oxidizing properties, as the additional oxygen can be released and used to react with other substances.

Chemokine (C-C motif) ligand 5, also known as RANTES (Regulated on Activation, Normal T cell Expressed and Secreted), is a chemokine that plays a crucial role in the immune system. It is a small signaling protein that attracts and activates immune cells, such as leukocytes, to the sites of infection or inflammation. Chemokine CCL5 binds to specific receptors on the surface of target cells, including CCR1, CCR3, and CCR5, and triggers a cascade of intracellular signaling events that result in cell migration and activation.

Chemokine CCL5 is involved in various physiological and pathological processes, such as wound healing, immune surveillance, and inflammation. It has been implicated in the pathogenesis of several diseases, including HIV infection, rheumatoid arthritis, multiple sclerosis, and cancer. In HIV infection, Chemokine CCL5 can bind to and inhibit the entry of the virus into CD4+ T cells by blocking the interaction between the viral envelope protein gp120 and the chemokine receptor CCR5. However, in advanced stages of HIV infection, the virus may develop resistance to this inhibitory effect, leading to increased viral replication and disease progression.

Lymphokines are a type of cytokines that are produced and released by activated lymphocytes, a type of white blood cell, in response to an antigenic stimulation. They play a crucial role in the regulation of immune responses and inflammation. Lymphokines can mediate various biological activities such as chemotaxis, activation, proliferation, and differentiation of different immune cells including lymphocytes, monocytes, macrophages, and eosinophils. Examples of lymphokines include interleukins (ILs), interferons (IFNs), tumor necrosis factor (TNF), and colony-stimulating factors (CSFs).

Immunologic adjuvants are substances that are added to a vaccine to enhance the body's immune response to the antigens contained in the vaccine. They work by stimulating the immune system and promoting the production of antibodies and activating immune cells, such as T-cells and macrophages, which help to provide a stronger and more sustained immune response to the vaccine.

Immunologic adjuvants can be derived from various sources, including bacteria, viruses, and chemicals. Some common examples include aluminum salts (alum), oil-in-water emulsions (such as MF59), and bacterial components (such as lipopolysaccharide or LPS).

The use of immunologic adjuvants in vaccines can help to improve the efficacy of the vaccine, particularly for vaccines that contain weak or poorly immunogenic antigens. They can also help to reduce the amount of antigen needed in a vaccine, which can be beneficial for vaccines that are difficult or expensive to produce.

It's important to note that while adjuvants can enhance the immune response to a vaccine, they can also increase the risk of adverse reactions, such as inflammation and pain at the injection site. Therefore, the use of immunologic adjuvants must be carefully balanced against their potential benefits and risks.

An amino acid sequence is the specific order of amino acids in a protein or peptide molecule, formed by the linking of the amino group (-NH2) of one amino acid to the carboxyl group (-COOH) of another amino acid through a peptide bond. The sequence is determined by the genetic code and is unique to each type of protein or peptide. It plays a crucial role in determining the three-dimensional structure and function of proteins.

I must clarify that the term "Guinea Pigs" is not typically used in medical definitions. However, in colloquial or informal language, it may refer to people who are used as the first to try out a new medical treatment or drug. This is known as being a "test subject" or "in a clinical trial."

In the field of scientific research, particularly in studies involving animals, guinea pigs are small rodents that are often used as experimental subjects due to their size, cost-effectiveness, and ease of handling. They are not actually pigs from Guinea, despite their name's origins being unclear. However, they do not exactly fit the description of being used in human medical experiments.

Genetic transcription is the process by which the information in a strand of DNA is used to create a complementary RNA molecule. This process is the first step in gene expression, where the genetic code in DNA is converted into a form that can be used to produce proteins or functional RNAs.

During transcription, an enzyme called RNA polymerase binds to the DNA template strand and reads the sequence of nucleotide bases. As it moves along the template, it adds complementary RNA nucleotides to the growing RNA chain, creating a single-stranded RNA molecule that is complementary to the DNA template strand. Once transcription is complete, the RNA molecule may undergo further processing before it can be translated into protein or perform its functional role in the cell.

Transcription can be either "constitutive" or "regulated." Constitutive transcription occurs at a relatively constant rate and produces essential proteins that are required for basic cellular functions. Regulated transcription, on the other hand, is subject to control by various intracellular and extracellular signals, allowing cells to respond to changing environmental conditions or developmental cues.

"Specific Pathogen-Free (SPF)" is a term used to describe animals or organisms that are raised and maintained in a controlled environment, free from specific pathogens (disease-causing agents) that could interfere with research outcomes or pose a risk to human or animal health. The "specific" part of the term refers to the fact that the exclusion of pathogens is targeted to those that are relevant to the particular organism or research being conducted.

To maintain an SPF status, animals are typically housed in specialized facilities with strict biosecurity measures, such as air filtration systems, quarantine procedures, and rigorous sanitation protocols. They are usually bred and raised in isolation from other animals, and their health status is closely monitored to ensure that they remain free from specific pathogens.

It's important to note that SPF does not necessarily mean "germ-free" or "sterile," as some microorganisms may still be present in the environment or on the animals themselves, even in an SPF facility. Instead, it means that the animals are free from specific pathogens that have been identified and targeted for exclusion.

In summary, Specific Pathogen-Free Organisms refer to animals or organisms that are raised and maintained in a controlled environment, free from specific disease-causing agents that are relevant to the research being conducted or human/animal health.

Interleukin-12 (IL-12) is a heterodimeric cytokine composed of two subunits, p35 and p40. IL-12 subunit p40 is a 40 kDa protein that forms the alpha chain of the IL-12 heterodimer. It can also form a homodimer called IL-23 with another subunit, p19, which has distinct biological activities from IL-12.

IL-12 plays an essential role in the differentiation of naive CD4+ T cells into Th1 cells and the production of interferon-gamma (IFN-γ). It is produced primarily by activated dendritic cells, macrophages, and neutrophils in response to bacterial or viral infections. IL-12 p40 subunit is involved in the binding of IL-12 to its receptor, which consists of two chains, IL-12Rβ1 and IL-12Rβ2.

Abnormalities in IL-12 signaling have been implicated in various diseases, including autoimmune disorders, chronic infections, and cancer. Therefore, IL-12 p40 subunit has become a target for therapeutic interventions in these conditions.

Transfection is a term used in molecular biology that refers to the process of deliberately introducing foreign genetic material (DNA, RNA or artificial gene constructs) into cells. This is typically done using chemical or physical methods, such as lipofection or electroporation. Transfection is widely used in research and medical settings for various purposes, including studying gene function, producing proteins, developing gene therapies, and creating genetically modified organisms. It's important to note that transfection is different from transduction, which is the process of introducing genetic material into cells using viruses as vectors.

'DBA' is an abbreviation for 'Database of Genotypes and Phenotypes,' but in the context of "Inbred DBA mice," it refers to a specific strain of laboratory mice that have been inbred for many generations. The DBA strain is one of the oldest inbred strains, and it was established in 1909 by C.C. Little at the Bussey Institute of Harvard University.

The "Inbred DBA" mice are genetically identical mice that have been produced by brother-sister matings for more than 20 generations. This extensive inbreeding results in a homozygous population, where all members of the strain have the same genetic makeup. The DBA strain is further divided into several sub-strains, including DBA/1, DBA/2, and DBA/J, among others.

DBA mice are known for their black coat color, which can fade to gray with age, and they exhibit a range of phenotypic traits that make them useful for research purposes. For example, DBA mice have a high incidence of retinal degeneration, making them a valuable model for studying eye diseases. They also show differences in behavior, immune response, and susceptibility to various diseases compared to other inbred strains.

In summary, "Inbred DBA" mice are a specific strain of laboratory mice that have been inbred for many generations, resulting in a genetically identical population with distinct phenotypic traits. They are widely used in biomedical research to study various diseases and biological processes.

Immunoglobulin G (IgG) is a type of antibody, which is a protective protein produced by the immune system in response to foreign substances like bacteria or viruses. IgG is the most abundant type of antibody in human blood, making up about 75-80% of all antibodies. It is found in all body fluids and plays a crucial role in fighting infections caused by bacteria, viruses, and toxins.

IgG has several important functions:

1. Neutralization: IgG can bind to the surface of bacteria or viruses, preventing them from attaching to and infecting human cells.
2. Opsonization: IgG coats the surface of pathogens, making them more recognizable and easier for immune cells like neutrophils and macrophages to phagocytose (engulf and destroy) them.
3. Complement activation: IgG can activate the complement system, a group of proteins that work together to help eliminate pathogens from the body. Activation of the complement system leads to the formation of the membrane attack complex, which creates holes in the cell membranes of bacteria, leading to their lysis (destruction).
4. Antibody-dependent cellular cytotoxicity (ADCC): IgG can bind to immune cells like natural killer (NK) cells and trigger them to release substances that cause target cells (such as virus-infected or cancerous cells) to undergo apoptosis (programmed cell death).
5. Immune complex formation: IgG can form immune complexes with antigens, which can then be removed from the body through various mechanisms, such as phagocytosis by immune cells or excretion in urine.

IgG is a critical component of adaptive immunity and provides long-lasting protection against reinfection with many pathogens. It has four subclasses (IgG1, IgG2, IgG3, and IgG4) that differ in their structure, function, and distribution in the body.

Orphan nuclear receptors are a subfamily of nuclear receptor proteins that are classified as "orphans" because their specific endogenous ligands (natural activating molecules) have not yet been identified. These receptors are still functional transcription factors, which means they can bind to specific DNA sequences and regulate the expression of target genes when activated by a ligand. However, in the case of orphan nuclear receptors, the identity of these ligands remains unknown or unconfirmed.

These receptors play crucial roles in various biological processes, including development, metabolism, and homeostasis. Some orphan nuclear receptors have been found to bind to synthetic ligands (man-made molecules), which has led to the development of potential therapeutic agents for various diseases. Over time, as research progresses, some orphan nuclear receptors may eventually have their endogenous ligands identified and be reclassified as non-orphan nuclear receptors.

Antibodies are proteins produced by the immune system in response to the presence of a foreign substance, such as a bacterium or virus. They are capable of identifying and binding to specific antigens (foreign substances) on the surface of these invaders, marking them for destruction by other immune cells. Antibodies are also known as immunoglobulins and come in several different types, including IgA, IgD, IgE, IgG, and IgM, each with a unique function in the immune response. They are composed of four polypeptide chains, two heavy chains and two light chains, that are held together by disulfide bonds. The variable regions of the heavy and light chains form the antigen-binding site, which is specific to a particular antigen.

Bone marrow is the spongy tissue found inside certain bones in the body, such as the hips, thighs, and vertebrae. It is responsible for producing blood-forming cells, including red blood cells, white blood cells, and platelets. There are two types of bone marrow: red marrow, which is involved in blood cell production, and yellow marrow, which contains fatty tissue.

Red bone marrow contains hematopoietic stem cells, which can differentiate into various types of blood cells. These stem cells continuously divide and mature to produce new blood cells that are released into the circulation. Red blood cells carry oxygen throughout the body, white blood cells help fight infections, and platelets play a crucial role in blood clotting.

Bone marrow also serves as a site for immune cell development and maturation. It contains various types of immune cells, such as lymphocytes, macrophages, and dendritic cells, which help protect the body against infections and diseases.

Abnormalities in bone marrow function can lead to several medical conditions, including anemia, leukopenia, thrombocytopenia, and various types of cancer, such as leukemia and multiple myeloma. Bone marrow aspiration and biopsy are common diagnostic procedures used to evaluate bone marrow health and function.

Granulocytes are a type of white blood cell that plays a crucial role in the body's immune system. They are called granulocytes because they contain small granules in their cytoplasm, which are filled with various enzymes and proteins that help them fight off infections and destroy foreign substances.

There are three types of granulocytes: neutrophils, eosinophils, and basophils. Neutrophils are the most abundant type and are primarily responsible for fighting bacterial infections. Eosinophils play a role in defending against parasitic infections and regulating immune responses. Basophils are involved in inflammatory reactions and allergic responses.

Granulocytes are produced in the bone marrow and released into the bloodstream, where they circulate and patrol for any signs of infection or foreign substances. When they encounter a threat, they quickly move to the site of infection or injury and release their granules to destroy the invading organisms or substances.

Abnormal levels of granulocytes in the blood can indicate an underlying medical condition, such as an infection, inflammation, or a bone marrow disorder.

A cell membrane, also known as the plasma membrane, is a thin semi-permeable phospholipid bilayer that surrounds all cells in animals, plants, and microorganisms. It functions as a barrier to control the movement of substances in and out of the cell, allowing necessary molecules such as nutrients, oxygen, and signaling molecules to enter while keeping out harmful substances and waste products. The cell membrane is composed mainly of phospholipids, which have hydrophilic (water-loving) heads and hydrophobic (water-fearing) tails. This unique structure allows the membrane to be flexible and fluid, yet selectively permeable. Additionally, various proteins are embedded in the membrane that serve as channels, pumps, receptors, and enzymes, contributing to the cell's overall functionality and communication with its environment.

"Salmonella enterica" serovar "Typhimurium" is a subspecies of the bacterial species Salmonella enterica, which is a gram-negative, facultatively anaerobic, rod-shaped bacterium. It is a common cause of foodborne illness in humans and animals worldwide. The bacteria can be found in a variety of sources, including contaminated food and water, raw meat, poultry, eggs, and dairy products.

The infection caused by Salmonella Typhimurium is typically self-limiting and results in gastroenteritis, which is characterized by symptoms such as diarrhea, abdominal cramps, fever, and vomiting. However, in some cases, the infection can spread to other parts of the body and cause more severe illness, particularly in young children, older adults, and people with weakened immune systems.

Salmonella Typhimurium is a major public health concern due to its ability to cause outbreaks of foodborne illness, as well as its potential to develop antibiotic resistance. Proper food handling, preparation, and storage practices can help prevent the spread of Salmonella Typhimurium and other foodborne pathogens.

Fibroblasts are specialized cells that play a critical role in the body's immune response and wound healing process. They are responsible for producing and maintaining the extracellular matrix (ECM), which is the non-cellular component present within all tissues and organs, providing structural support and biochemical signals for surrounding cells.

Fibroblasts produce various ECM proteins such as collagens, elastin, fibronectin, and laminins, forming a complex network of fibers that give tissues their strength and flexibility. They also help in the regulation of tissue homeostasis by controlling the turnover of ECM components through the process of remodeling.

In response to injury or infection, fibroblasts become activated and start to proliferate rapidly, migrating towards the site of damage. Here, they participate in the inflammatory response, releasing cytokines and chemokines that attract immune cells to the area. Additionally, they deposit new ECM components to help repair the damaged tissue and restore its functionality.

Dysregulation of fibroblast activity has been implicated in several pathological conditions, including fibrosis (excessive scarring), cancer (where they can contribute to tumor growth and progression), and autoimmune diseases (such as rheumatoid arthritis).

Protein binding, in the context of medical and biological sciences, refers to the interaction between a protein and another molecule (known as the ligand) that results in a stable complex. This process is often reversible and can be influenced by various factors such as pH, temperature, and concentration of the involved molecules.

In clinical chemistry, protein binding is particularly important when it comes to drugs, as many of them bind to proteins (especially albumin) in the bloodstream. The degree of protein binding can affect a drug's distribution, metabolism, and excretion, which in turn influence its therapeutic effectiveness and potential side effects.

Protein-bound drugs may be less available for interaction with their target tissues, as only the unbound or "free" fraction of the drug is active. Therefore, understanding protein binding can help optimize dosing regimens and minimize adverse reactions.

Medical Definition:

Matrix metalloproteinase 9 (MMP-9), also known as gelatinase B or 92 kDa type IV collagenase, is a member of the matrix metalloproteinase family. These enzymes are involved in degrading and remodeling the extracellular matrix (ECM) components, playing crucial roles in various physiological and pathological processes such as wound healing, tissue repair, and tumor metastasis.

MMP-9 is secreted as an inactive zymogen and activated upon removal of its propeptide domain. It can degrade several ECM proteins, including type IV collagen, elastin, fibronectin, and gelatin. MMP-9 has been implicated in numerous diseases, such as cancer, rheumatoid arthritis, neurological disorders, and cardiovascular diseases. Its expression is regulated at the transcriptional, translational, and post-translational levels, and its activity can be controlled by endogenous inhibitors called tissue inhibitors of metalloproteinases (TIMPs).

Antigen-presenting cells (APCs) are a group of specialized cells in the immune system that play a critical role in initiating and regulating immune responses. They have the ability to engulf, process, and present antigens (molecules derived from pathogens or other foreign substances) on their surface in conjunction with major histocompatibility complex (MHC) molecules. This presentation of antigens allows APCs to activate T cells, which are crucial for adaptive immunity.

There are several types of APCs, including:

1. Dendritic cells (DCs): These are the most potent and professional APCs, found in various tissues throughout the body. DCs can capture antigens from their environment, process them, and migrate to lymphoid organs where they present antigens to T cells.
2. Macrophages: These large phagocytic cells are found in many tissues and play a role in both innate and adaptive immunity. They can engulf and digest pathogens, then present processed antigens on their MHC class II molecules to activate CD4+ T helper cells.
3. B cells: These are primarily responsible for humoral immune responses by producing antibodies against antigens. When activated, B cells can also function as APCs and present antigens on their MHC class II molecules to CD4+ T cells.

The interaction between APCs and T cells is critical for the development of an effective immune response against pathogens or other foreign substances. This process helps ensure that the immune system can recognize and eliminate threats while minimizing damage to healthy tissues.

Necrosis is the premature death of cells or tissues due to damage or injury, such as from infection, trauma, infarction (lack of blood supply), or toxic substances. It's a pathological process that results in the uncontrolled and passive degradation of cellular components, ultimately leading to the release of intracellular contents into the extracellular space. This can cause local inflammation and may lead to further tissue damage if not treated promptly.

There are different types of necrosis, including coagulative, liquefactive, caseous, fat, fibrinoid, and gangrenous necrosis, each with distinct histological features depending on the underlying cause and the affected tissues or organs.

Immunoenzyme techniques are a group of laboratory methods used in immunology and clinical chemistry that combine the specificity of antibody-antigen reactions with the sensitivity and amplification capabilities of enzyme reactions. These techniques are primarily used for the detection, quantitation, or identification of various analytes (such as proteins, hormones, drugs, viruses, or bacteria) in biological samples.

In immunoenzyme techniques, an enzyme is linked to an antibody or antigen, creating a conjugate. This conjugate then interacts with the target analyte in the sample, forming an immune complex. The presence and amount of this immune complex can be visualized or measured by detecting the enzymatic activity associated with it.

There are several types of immunoenzyme techniques, including:

1. Enzyme-linked Immunosorbent Assay (ELISA): A widely used method for detecting and quantifying various analytes in a sample. In ELISA, an enzyme is attached to either the capture antibody or the detection antibody. After the immune complex formation, a substrate is added that reacts with the enzyme, producing a colored product that can be measured spectrophotometrically.
2. Immunoblotting (Western blot): A method used for detecting specific proteins in a complex mixture, such as a protein extract from cells or tissues. In this technique, proteins are separated by gel electrophoresis and transferred to a membrane, where they are probed with an enzyme-conjugated antibody directed against the target protein.
3. Immunohistochemistry (IHC): A method used for detecting specific antigens in tissue sections or cells. In IHC, an enzyme-conjugated primary or secondary antibody is applied to the sample, and the presence of the antigen is visualized using a chromogenic substrate that produces a colored product at the site of the antigen-antibody interaction.
4. Immunofluorescence (IF): A method used for detecting specific antigens in cells or tissues by employing fluorophore-conjugated antibodies. The presence of the antigen is visualized using a fluorescence microscope.
5. Enzyme-linked immunosorbent assay (ELISA): A method used for detecting and quantifying specific antigens or antibodies in liquid samples, such as serum or culture supernatants. In ELISA, an enzyme-conjugated detection antibody is added after the immune complex formation, and a substrate is added that reacts with the enzyme to produce a colored product that can be measured spectrophotometrically.

These techniques are widely used in research and diagnostic laboratories for various applications, including protein characterization, disease diagnosis, and monitoring treatment responses.

"Leishmania major" is a species of parasitic protozoan that causes cutaneous leishmaniasis, a type of disease transmitted through the bite of infected female sandflies. The organism's life cycle involves two main stages: the promastigote stage, which develops in the sandfly vector and is infective to mammalian hosts; and the amastigote stage, which resides inside host cells such as macrophages and dendritic cells, where it replicates.

The disease caused by L. major typically results in skin ulcers or lesions that can take several months to heal and may leave permanent scars. While not usually life-threatening, cutaneous leishmaniasis can cause significant disfigurement and psychological distress, particularly when it affects the face. In addition, people with weakened immune systems, such as those with HIV/AIDS or those undergoing immunosuppressive therapy, may be at risk of developing more severe forms of the disease.

L. major is found primarily in the Old World, including parts of North Africa, the Middle East, and Central Asia. It is transmitted by various species of sandflies belonging to the genus Phlebotomus. Preventive measures include using insect repellent, wearing protective clothing, and reducing outdoor activities during peak sandfly feeding times.

Sprague-Dawley rats are a strain of albino laboratory rats that are widely used in scientific research. They were first developed by researchers H.H. Sprague and R.C. Dawley in the early 20th century, and have since become one of the most commonly used rat strains in biomedical research due to their relatively large size, ease of handling, and consistent genetic background.

Sprague-Dawley rats are outbred, which means that they are genetically diverse and do not suffer from the same limitations as inbred strains, which can have reduced fertility and increased susceptibility to certain diseases. They are also characterized by their docile nature and low levels of aggression, making them easier to handle and study than some other rat strains.

These rats are used in a wide variety of research areas, including toxicology, pharmacology, nutrition, cancer, and behavioral studies. Because they are genetically diverse, Sprague-Dawley rats can be used to model a range of human diseases and conditions, making them an important tool in the development of new drugs and therapies.

Bacterial proteins are a type of protein that are produced by bacteria as part of their structural or functional components. These proteins can be involved in various cellular processes, such as metabolism, DNA replication, transcription, and translation. They can also play a role in bacterial pathogenesis, helping the bacteria to evade the host's immune system, acquire nutrients, and multiply within the host.

Bacterial proteins can be classified into different categories based on their function, such as:

1. Enzymes: Proteins that catalyze chemical reactions in the bacterial cell.
2. Structural proteins: Proteins that provide structural support and maintain the shape of the bacterial cell.
3. Signaling proteins: Proteins that help bacteria to communicate with each other and coordinate their behavior.
4. Transport proteins: Proteins that facilitate the movement of molecules across the bacterial cell membrane.
5. Toxins: Proteins that are produced by pathogenic bacteria to damage host cells and promote infection.
6. Surface proteins: Proteins that are located on the surface of the bacterial cell and interact with the environment or host cells.

Understanding the structure and function of bacterial proteins is important for developing new antibiotics, vaccines, and other therapeutic strategies to combat bacterial infections.

A dose-response relationship in immunology refers to the quantitative relationship between the dose or amount of an antigen (a substance that triggers an immune response) and the magnitude or strength of the resulting immune response. Generally, as the dose of an antigen increases, the intensity and/or duration of the immune response also increase, up to a certain point. This relationship helps in determining the optimal dosage for vaccines and immunotherapies, ensuring sufficient immune activation while minimizing potential adverse effects.

Oxidation-Reduction (redox) reactions are a type of chemical reaction involving a transfer of electrons between two species. The substance that loses electrons in the reaction is oxidized, and the substance that gains electrons is reduced. Oxidation and reduction always occur together in a redox reaction, hence the term "oxidation-reduction."

In biological systems, redox reactions play a crucial role in many cellular processes, including energy production, metabolism, and signaling. The transfer of electrons in these reactions is often facilitated by specialized molecules called electron carriers, such as nicotinamide adenine dinucleotide (NAD+/NADH) and flavin adenine dinucleotide (FAD/FADH2).

The oxidation state of an element in a compound is a measure of the number of electrons that have been gained or lost relative to its neutral state. In redox reactions, the oxidation state of one or more elements changes as they gain or lose electrons. The substance that is oxidized has a higher oxidation state, while the substance that is reduced has a lower oxidation state.

Overall, oxidation-reduction reactions are fundamental to the functioning of living organisms and are involved in many important biological processes.

Chemokine receptors are a type of G protein-coupled receptor (GPCR) that bind to chemokines, which are small signaling proteins involved in immune cell trafficking and inflammation. These receptors play a crucial role in the regulation of immune responses, hematopoiesis, and development. Chemokine receptors are expressed on the surface of various cells, including leukocytes, endothelial cells, and fibroblasts. Upon binding to their respective chemokines, these receptors activate intracellular signaling pathways that lead to cell migration, activation, or proliferation. There are several subfamilies of chemokine receptors, including CXCR, CCR, CX3CR, and XCR, each with distinct specificities for different chemokines. Dysregulation of chemokine receptor signaling has been implicated in various pathological conditions, such as autoimmune diseases, cancer, and viral infections.

Lipid metabolism is the process by which the body breaks down and utilizes lipids (fats) for various functions, such as energy production, cell membrane formation, and hormone synthesis. This complex process involves several enzymes and pathways that regulate the digestion, absorption, transport, storage, and consumption of fats in the body.

The main types of lipids involved in metabolism include triglycerides, cholesterol, phospholipids, and fatty acids. The breakdown of these lipids begins in the digestive system, where enzymes called lipases break down dietary fats into smaller molecules called fatty acids and glycerol. These molecules are then absorbed into the bloodstream and transported to the liver, which is the main site of lipid metabolism.

In the liver, fatty acids may be further broken down for energy production or used to synthesize new lipids. Excess fatty acids may be stored as triglycerides in specialized cells called adipocytes (fat cells) for later use. Cholesterol is also metabolized in the liver, where it may be used to synthesize bile acids, steroid hormones, and other important molecules.

Disorders of lipid metabolism can lead to a range of health problems, including obesity, diabetes, cardiovascular disease, and non-alcoholic fatty liver disease (NAFLD). These conditions may be caused by genetic factors, lifestyle habits, or a combination of both. Proper diagnosis and management of lipid metabolism disorders typically involves a combination of dietary changes, exercise, and medication.

Chemokines are a family of small proteins that are involved in immune responses and inflammation. They mediate the chemotaxis (directed migration) of various cells, including leukocytes (white blood cells). Chemokines are classified into four major subfamilies based on the arrangement of conserved cysteine residues near the amino terminus: CXC, CC, C, and CX3C.

CC chemokines, also known as β-chemokines, are characterized by the presence of two adjacent cysteine residues near their N-terminal end. There are 27 known human CC chemokines, including MCP-1 (monocyte chemoattractant protein-1), RANTES (regulated on activation, normal T cell expressed and secreted), and eotaxin.

CC chemokines play important roles in the recruitment of immune cells to sites of infection or injury, as well as in the development and maintenance of immune responses. They bind to specific G protein-coupled receptors (GPCRs) on the surface of target cells, leading to the activation of intracellular signaling pathways that regulate cell migration, proliferation, and survival.

Dysregulation of CC chemokines and their receptors has been implicated in various inflammatory and autoimmune diseases, as well as in cancer. Therefore, targeting CC chemokine-mediated signaling pathways has emerged as a promising therapeutic strategy for the treatment of these conditions.

Confocal microscopy is a powerful imaging technique used in medical and biological research to obtain high-resolution, contrast-rich images of thick samples. This super-resolution technology provides detailed visualization of cellular structures and processes at various depths within a specimen.

In confocal microscopy, a laser beam focused through a pinhole illuminates a small spot within the sample. The emitted fluorescence or reflected light from this spot is then collected by a detector, passing through a second pinhole that ensures only light from the focal plane reaches the detector. This process eliminates out-of-focus light, resulting in sharp images with improved contrast compared to conventional widefield microscopy.

By scanning the laser beam across the sample in a raster pattern and collecting fluorescence at each point, confocal microscopy generates optical sections of the specimen. These sections can be combined to create three-dimensional reconstructions, allowing researchers to study cellular architecture and interactions within complex tissues.

Confocal microscopy has numerous applications in medical research, including studying protein localization, tracking intracellular dynamics, analyzing cell morphology, and investigating disease mechanisms at the cellular level. Additionally, it is widely used in clinical settings for diagnostic purposes, such as analyzing skin lesions or detecting pathogens in patient samples.

Gene expression profiling is a laboratory technique used to measure the activity (expression) of thousands of genes at once. This technique allows researchers and clinicians to identify which genes are turned on or off in a particular cell, tissue, or organism under specific conditions, such as during health, disease, development, or in response to various treatments.

The process typically involves isolating RNA from the cells or tissues of interest, converting it into complementary DNA (cDNA), and then using microarray or high-throughput sequencing technologies to determine which genes are expressed and at what levels. The resulting data can be used to identify patterns of gene expression that are associated with specific biological states or processes, providing valuable insights into the underlying molecular mechanisms of diseases and potential targets for therapeutic intervention.

In recent years, gene expression profiling has become an essential tool in various fields, including cancer research, drug discovery, and personalized medicine, where it is used to identify biomarkers of disease, predict patient outcomes, and guide treatment decisions.

Acetylmuramyl-Alanyl-Isoglutamine is a chemical compound that is a component of bacterial cell walls. It is also known as N-acetylmuramic acid-L-alanine-γ-D-glutamyl-meso-diaminopimelic acid, which is its more detailed and complete chemical name.

This compound is a key building block of peptidoglycan, a complex polymer that provides structural rigidity to bacterial cell walls. Specifically, Acetylmuramyl-Alanyl-Isoglutamine is a part of the peptide subunit that links individual peptidoglycan strands together, forming a cross-linked network that helps protect bacteria from external stresses and osmotic pressure.

In addition to its structural role, Acetylmuramyl-Alanyl-Isoglutamine has been shown to have immunostimulatory properties, and it is being investigated as a potential vaccine adjuvant to enhance the immune response to other antigens.

Fluorescence microscopy is a type of microscopy that uses fluorescent dyes or proteins to highlight and visualize specific components within a sample. In this technique, the sample is illuminated with high-energy light, typically ultraviolet (UV) or blue light, which excites the fluorescent molecules causing them to emit lower-energy, longer-wavelength light, usually visible light in the form of various colors. This emitted light is then collected by the microscope and detected to produce an image.

Fluorescence microscopy has several advantages over traditional brightfield microscopy, including the ability to visualize specific structures or molecules within a complex sample, increased sensitivity, and the potential for quantitative analysis. It is widely used in various fields of biology and medicine, such as cell biology, neuroscience, and pathology, to study the structure, function, and interactions of cells and proteins.

There are several types of fluorescence microscopy techniques, including widefield fluorescence microscopy, confocal microscopy, two-photon microscopy, and total internal reflection fluorescence (TIRF) microscopy, each with its own strengths and limitations. These techniques can provide valuable insights into the behavior of cells and proteins in health and disease.

Surface antigens are molecules found on the surface of cells that can be recognized by the immune system as being foreign or different from the host's own cells. Antigens are typically proteins or polysaccharides that are capable of stimulating an immune response, leading to the production of antibodies and activation of immune cells such as T-cells.

Surface antigens are important in the context of infectious diseases because they allow the immune system to identify and target infected cells for destruction. For example, viruses and bacteria often display surface antigens that are distinct from those found on host cells, allowing the immune system to recognize and attack them. In some cases, these surface antigens can also be used as targets for vaccines or other immunotherapies.

In addition to their role in infectious diseases, surface antigens are also important in the context of cancer. Tumor cells often display abnormal surface antigens that differ from those found on normal cells, allowing the immune system to potentially recognize and attack them. However, tumors can also develop mechanisms to evade the immune system, making it difficult to mount an effective response.

Overall, understanding the properties and behavior of surface antigens is crucial for developing effective immunotherapies and vaccines against infectious diseases and cancer.

Endocytosis is the process by which cells absorb substances from their external environment by engulfing them in membrane-bound structures, resulting in the formation of intracellular vesicles. This mechanism allows cells to take up large molecules, such as proteins and lipids, as well as small particles, like bacteria and viruses. There are two main types of endocytosis: phagocytosis (cell eating) and pinocytosis (cell drinking). Phagocytosis involves the engulfment of solid particles, while pinocytosis deals with the uptake of fluids and dissolved substances. Other specialized forms of endocytosis include receptor-mediated endocytosis and caveolae-mediated endocytosis, which allow for the specific internalization of molecules through the interaction with cell surface receptors.

Immunophenotyping is a medical laboratory technique used to identify and classify cells, usually in the context of hematologic (blood) disorders and malignancies (cancers), based on their surface or intracellular expression of various proteins and antigens. This technique utilizes specific antibodies tagged with fluorochromes, which bind to the target antigens on the cell surface or within the cells. The labeled cells are then analyzed using flow cytometry, allowing for the detection and quantification of multiple antigenic markers simultaneously.

Immunophenotyping helps in understanding the distribution of different cell types, their subsets, and activation status, which can be crucial in diagnosing various hematological disorders, immunodeficiencies, and distinguishing between different types of leukemias, lymphomas, and other malignancies. Additionally, it can also be used to monitor the progression of diseases, evaluate the effectiveness of treatments, and detect minimal residual disease (MRD) during follow-up care.

Host-pathogen interactions refer to the complex and dynamic relationship between a living organism (the host) and a disease-causing agent (the pathogen). This interaction can involve various molecular, cellular, and physiological processes that occur between the two entities. The outcome of this interaction can determine whether the host will develop an infection or not, as well as the severity and duration of the illness.

During host-pathogen interactions, the pathogen may release virulence factors that allow it to evade the host's immune system, colonize tissues, and obtain nutrients for its survival and replication. The host, in turn, may mount an immune response to recognize and eliminate the pathogen, which can involve various mechanisms such as inflammation, phagocytosis, and the production of antimicrobial agents.

Understanding the intricacies of host-pathogen interactions is crucial for developing effective strategies to prevent and treat infectious diseases. This knowledge can help identify new targets for therapeutic interventions, inform vaccine design, and guide public health policies to control the spread of infectious agents.

The peritoneum is the serous membrane that lines the abdominal cavity and covers the abdominal organs. It is composed of a mesothelial cell monolayer supported by a thin, loose connective tissue. The peritoneum has two layers: the parietal peritoneum, which lines the abdominal wall, and the visceral peritoneum, which covers the organs.

The potential space between these two layers is called the peritoneal cavity, which contains a small amount of serous fluid that allows for the smooth movement of the organs within the cavity. The peritoneum plays an important role in the absorption and secretion of fluids and electrolytes, as well as providing a surface for the circulation of immune cells.

In addition, it also provides a route for the spread of infection or malignant cells throughout the abdominal cavity, known as peritonitis. The peritoneum is highly vascularized and innervated, making it sensitive to pain and distention.

An antigen-antibody complex is a type of immune complex that forms when an antibody binds to a specific antigen. An antigen is any substance that triggers an immune response, while an antibody is a protein produced by the immune system to neutralize or destroy foreign substances like antigens.

When an antibody binds to an antigen, it forms a complex that can be either soluble or insoluble. Soluble complexes are formed when the antigen is small and can move freely through the bloodstream. Insoluble complexes, on the other hand, are formed when the antigen is too large to move freely, such as when it is part of a bacterium or virus.

The formation of antigen-antibody complexes plays an important role in the immune response. Once formed, these complexes can be recognized and cleared by other components of the immune system, such as phagocytes, which help to prevent further damage to the body. However, in some cases, the formation of large numbers of antigen-antibody complexes can lead to inflammation and tissue damage, contributing to the development of certain autoimmune diseases.

Myeloid cells are a type of immune cell that originate from the bone marrow. They develop from hematopoietic stem cells, which can differentiate into various types of blood cells. Myeloid cells include monocytes, macrophages, granulocytes (such as neutrophils, eosinophils, and basophils), dendritic cells, and mast cells. These cells play important roles in the immune system, such as defending against pathogens, modulating inflammation, and participating in tissue repair and remodeling.

Myeloid cell development is a tightly regulated process that involves several stages of differentiation, including the commitment to the myeloid lineage, proliferation, and maturation into specific subtypes. Dysregulation of myeloid cell development or function can contribute to various diseases, such as infections, cancer, and autoimmune disorders.

DNA primers are short single-stranded DNA molecules that serve as a starting point for DNA synthesis. They are typically used in laboratory techniques such as the polymerase chain reaction (PCR) and DNA sequencing. The primer binds to a complementary sequence on the DNA template through base pairing, providing a free 3'-hydroxyl group for the DNA polymerase enzyme to add nucleotides and synthesize a new strand of DNA. This allows for specific and targeted amplification or analysis of a particular region of interest within a larger DNA molecule.

CCR5 (C-C chemokine receptor type 5) is a type of protein found on the surface of certain white blood cells, including T-cells, macrophages, and dendritic cells. It belongs to the family of G protein-coupled receptors, which are involved in various cellular responses.

CCR5 acts as a co-receptor for HIV (Human Immunodeficiency Virus) entry into host cells, along with CD4. The virus binds to both CCR5 and CD4, leading to fusion of the viral and cell membranes and subsequent infection of the cell.

Individuals who have a genetic mutation that prevents CCR5 from functioning are resistant to HIV infection, highlighting its importance in the viral life cycle. Additionally, CCR5 antagonists have been developed as potential therapeutic agents for the treatment of HIV infection.

Lectins are a type of proteins that bind specifically to carbohydrates and have been found in various plant and animal sources. They play important roles in biological recognition events, such as cell-cell adhesion, and can also be involved in the immune response. Some lectins can agglutinate certain types of cells or precipitate glycoproteins, while others may have a more direct effect on cellular processes. In some cases, lectins from plants can cause adverse effects in humans if ingested, such as digestive discomfort or allergic reactions.

Lipoproteins are complex particles composed of multiple proteins and lipids (fats) that play a crucial role in the transport and metabolism of fat molecules in the body. They consist of an outer shell of phospholipids, free cholesterols, and apolipoproteins, enclosing a core of triglycerides and cholesteryl esters.

There are several types of lipoproteins, including:

1. Chylomicrons: These are the largest lipoproteins and are responsible for transporting dietary lipids from the intestines to other parts of the body.
2. Very-low-density lipoproteins (VLDL): Produced by the liver, VLDL particles carry triglycerides to peripheral tissues for energy storage or use.
3. Low-density lipoproteins (LDL): Often referred to as "bad cholesterol," LDL particles transport cholesterol from the liver to cells throughout the body. High levels of LDL in the blood can lead to plaque buildup in artery walls and increase the risk of heart disease.
4. High-density lipoproteins (HDL): Known as "good cholesterol," HDL particles help remove excess cholesterol from cells and transport it back to the liver for excretion or recycling. Higher levels of HDL are associated with a lower risk of heart disease.

Understanding lipoproteins and their roles in the body is essential for assessing cardiovascular health and managing risks related to heart disease and stroke.

Hematopoietic stem cells (HSCs) are immature, self-renewing cells that give rise to all the mature blood and immune cells in the body. They are capable of both producing more hematopoietic stem cells (self-renewal) and differentiating into early progenitor cells that eventually develop into red blood cells, white blood cells, and platelets. HSCs are found in the bone marrow, umbilical cord blood, and peripheral blood. They have the ability to repair damaged tissues and offer significant therapeutic potential for treating various diseases, including hematological disorders, genetic diseases, and cancer.

'Immune sera' refers to the serum fraction of blood that contains antibodies produced in response to an antigenic stimulus, such as a vaccine or an infection. These antibodies are proteins known as immunoglobulins, which are secreted by B cells (a type of white blood cell) and can recognize and bind to specific antigens. Immune sera can be collected from an immunized individual and used as a source of passive immunity to protect against infection or disease. It is often used in research and diagnostic settings to identify or measure the presence of specific antigens or antibodies.

Leishmaniasis is a complex of diseases caused by the protozoan parasites of the Leishmania species, which are transmitted to humans through the bite of infected female phlebotomine sandflies. The disease presents with a variety of clinical manifestations, depending upon the Leishmania species involved and the host's immune response.

There are three main forms of leishmaniasis: cutaneous leishmaniasis (CL), mucocutaneous leishmaniasis (MCL), and visceral leishmaniasis (VL), also known as kala-azar. CL typically presents with skin ulcers, while MCL is characterized by the destruction of mucous membranes in the nose, mouth, and throat. VL, the most severe form, affects internal organs such as the spleen, liver, and bone marrow, causing symptoms like fever, weight loss, anemia, and enlarged liver and spleen.

Leishmaniasis is prevalent in many tropical and subtropical regions, including parts of Asia, Africa, South America, and southern Europe. The prevention strategies include using insect repellents, wearing protective clothing, and improving housing conditions to minimize exposure to sandflies. Effective treatment options are available for leishmaniasis, depending on the form and severity of the disease, geographical location, and the Leishmania species involved.

Inbred A mice are a strain of laboratory mice that have been produced by many generations of brother-sister matings. This results in a high degree of genetic similarity among individuals within the strain, making them useful for research purposes where a consistent genetic background is desired. The Inbred A strain is maintained through continued brother-sister mating. It's important to note that while these mice are called "Inbred A," the designation does not refer to any specific medical condition or characteristic. Instead, it refers to the breeding practices used to create and maintain this particular strain of laboratory mice.

Erythrocytes, also known as red blood cells (RBCs), are the most common type of blood cell in circulating blood in mammals. They are responsible for transporting oxygen from the lungs to the body's tissues and carbon dioxide from the tissues to the lungs.

Erythrocytes are formed in the bone marrow and have a biconcave shape, which allows them to fold and bend easily as they pass through narrow blood vessels. They do not have a nucleus or mitochondria, which makes them more flexible but also limits their ability to reproduce or repair themselves.

In humans, erythrocytes are typically disc-shaped and measure about 7 micrometers in diameter. They contain the protein hemoglobin, which binds to oxygen and gives blood its red color. The lifespan of an erythrocyte is approximately 120 days, after which it is broken down in the liver and spleen.

Abnormalities in erythrocyte count or function can lead to various medical conditions, such as anemia, polycythemia, and sickle cell disease.

Cell proliferation is the process by which cells increase in number, typically through the process of cell division. In the context of biology and medicine, it refers to the reproduction of cells that makes up living tissue, allowing growth, maintenance, and repair. It involves several stages including the transition from a phase of quiescence (G0 phase) to an active phase (G1 phase), DNA replication in the S phase, and mitosis or M phase, where the cell divides into two daughter cells.

Abnormal or uncontrolled cell proliferation is a characteristic feature of many diseases, including cancer, where deregulated cell cycle control leads to excessive and unregulated growth of cells, forming tumors that can invade surrounding tissues and metastasize to distant sites in the body.

Atherosclerotic plaque is a deposit of fatty (cholesterol and fat) substances, calcium, and other substances in the inner lining of an artery. This plaque buildup causes the artery to narrow and harden, reducing blood flow through the artery, which can lead to serious cardiovascular conditions such as coronary artery disease, angina, heart attack, or stroke. The process of atherosclerosis develops gradually over decades and can start in childhood.

CD4-positive T-lymphocytes, also known as CD4+ T cells or helper T cells, are a type of white blood cell that plays a crucial role in the immune response. They express the CD4 receptor on their surface and help coordinate the immune system's response to infectious agents such as viruses and bacteria.

CD4+ T cells recognize and bind to specific antigens presented by antigen-presenting cells, such as dendritic cells or macrophages. Once activated, they can differentiate into various subsets of effector cells, including Th1, Th2, Th17, and Treg cells, each with distinct functions in the immune response.

CD4+ T cells are particularly important in the immune response to HIV (human immunodeficiency virus), which targets and destroys these cells, leading to a weakened immune system and increased susceptibility to opportunistic infections. The number of CD4+ T cells is often used as a marker of disease progression in HIV infection, with lower counts indicating more advanced disease.

Promoter regions in genetics refer to specific DNA sequences located near the transcription start site of a gene. They serve as binding sites for RNA polymerase and various transcription factors that regulate the initiation of gene transcription. These regulatory elements help control the rate of transcription and, therefore, the level of gene expression. Promoter regions can be composed of different types of sequences, such as the TATA box and CAAT box, and their organization and composition can vary between different genes and species.

Lymph nodes are small, bean-shaped organs that are part of the immune system. They are found throughout the body, especially in the neck, armpits, groin, and abdomen. Lymph nodes filter lymph fluid, which carries waste and unwanted substances such as bacteria, viruses, and cancer cells. They contain white blood cells called lymphocytes that help fight infections and diseases by attacking and destroying the harmful substances found in the lymph fluid. When an infection or disease is present, lymph nodes may swell due to the increased number of immune cells and fluid accumulation as they work to fight off the invaders.

Cell death is the process by which cells cease to function and eventually die. There are several ways that cells can die, but the two most well-known and well-studied forms of cell death are apoptosis and necrosis.

Apoptosis is a programmed form of cell death that occurs as a normal and necessary process in the development and maintenance of healthy tissues. During apoptosis, the cell's DNA is broken down into small fragments, the cell shrinks, and the membrane around the cell becomes fragmented, allowing the cell to be easily removed by phagocytic cells without causing an inflammatory response.

Necrosis, on the other hand, is a form of cell death that occurs as a result of acute tissue injury or overwhelming stress. During necrosis, the cell's membrane becomes damaged and the contents of the cell are released into the surrounding tissue, causing an inflammatory response.

There are also other forms of cell death, such as autophagy, which is a process by which cells break down their own organelles and proteins to recycle nutrients and maintain energy homeostasis, and pyroptosis, which is a form of programmed cell death that occurs in response to infection and involves the activation of inflammatory caspases.

Cell death is an important process in many physiological and pathological processes, including development, tissue homeostasis, and disease. Dysregulation of cell death can contribute to the development of various diseases, including cancer, neurodegenerative disorders, and autoimmune diseases.

A biological marker, often referred to as a biomarker, is a measurable indicator that reflects the presence or severity of a disease state, or a response to a therapeutic intervention. Biomarkers can be found in various materials such as blood, tissues, or bodily fluids, and they can take many forms, including molecular, histologic, radiographic, or physiological measurements.

In the context of medical research and clinical practice, biomarkers are used for a variety of purposes, such as:

1. Diagnosis: Biomarkers can help diagnose a disease by indicating the presence or absence of a particular condition. For example, prostate-specific antigen (PSA) is a biomarker used to detect prostate cancer.
2. Monitoring: Biomarkers can be used to monitor the progression or regression of a disease over time. For instance, hemoglobin A1c (HbA1c) levels are monitored in diabetes patients to assess long-term blood glucose control.
3. Predicting: Biomarkers can help predict the likelihood of developing a particular disease or the risk of a negative outcome. For example, the presence of certain genetic mutations can indicate an increased risk for breast cancer.
4. Response to treatment: Biomarkers can be used to evaluate the effectiveness of a specific treatment by measuring changes in the biomarker levels before and after the intervention. This is particularly useful in personalized medicine, where treatments are tailored to individual patients based on their unique biomarker profiles.

It's important to note that for a biomarker to be considered clinically valid and useful, it must undergo rigorous validation through well-designed studies, including demonstrating sensitivity, specificity, reproducibility, and clinical relevance.

Gene expression regulation, enzymologic refers to the biochemical processes and mechanisms that control the transcription and translation of specific genes into functional proteins or enzymes. This regulation is achieved through various enzymatic activities that can either activate or repress gene expression at different levels, such as chromatin remodeling, transcription factor activation, mRNA processing, and protein degradation.

Enzymologic regulation of gene expression involves the action of specific enzymes that catalyze chemical reactions involved in these processes. For example, histone-modifying enzymes can alter the structure of chromatin to make genes more or less accessible for transcription, while RNA polymerase and its associated factors are responsible for transcribing DNA into mRNA. Additionally, various enzymes are involved in post-transcriptional modifications of mRNA, such as splicing, capping, and tailing, which can affect the stability and translation of the transcript.

Overall, the enzymologic regulation of gene expression is a complex and dynamic process that allows cells to respond to changes in their environment and maintain proper physiological function.

Macrophage Activation Syndrome (MAS) is a severe, life-threatening complication of certain inflammatory diseases, including rheumatic diseases such as systemic juvenile idiopathic arthritis (sJIA), adult-onset Still's disease (AOSD), and catastrophic antiphospholipid syndrome (CAPS). It is also known as hemophagocytic lymphohistiocytosis (HLH) or secondary HLH.

MAS is characterized by the uncontrolled activation and proliferation of macrophages, which are large white blood cells that play a crucial role in the immune system by engulfing and destroying foreign substances, microbes, and cancer cells. In MAS, these activated macrophages release high levels of inflammatory cytokines, leading to a hyperinflammatory state that can damage multiple organs, including the liver, spleen, kidneys, and central nervous system.

The symptoms of MAS include fever, fatigue, rash, lymphadenopathy (swollen lymph nodes), hepatosplenomegaly (enlarged liver and spleen), coagulopathy (bleeding disorders), and cytopenias (low blood cell counts). The diagnosis of MAS is based on clinical criteria, laboratory tests, and bone marrow aspiration findings. Treatment typically involves high-dose corticosteroids, immunosuppressive agents, and targeted therapies that modulate the immune system. In severe cases, stem cell transplantation may be necessary.

Purinergic P2X7 receptors are a type of ligand-gated ion channel that are activated by the binding of extracellular adenosine triphosphate (ATP) to the P2X7 receptor subunit. These receptors play important roles in various physiological and pathophysiological processes, including inflammation, immune response, pain perception, and cell death.

Upon activation of P2X7 receptors, there is an increase in membrane permeability to small cations such as Na+, K+, and Ca2+, which can lead to the depolarization of the cell membrane. Prolonged activation of these receptors can result in the formation of large pores that allow for the passage of larger molecules, including inflammatory mediators and even small proteins. This can ultimately lead to the induction of apoptosis or necrosis in certain cells.

P2X7 receptors are widely expressed in various tissues, including the brain, spinal cord, immune cells, and epithelial cells. In recent years, there has been growing interest in targeting P2X7 receptors for therapeutic purposes, particularly in the context of inflammatory diseases and chronic pain.

The synovial membrane, also known as the synovium, is the soft tissue that lines the inner surface of the capsule of a synovial joint, which is a type of joint that allows for smooth movement between bones. This membrane secretes synovial fluid, a viscous substance that lubricates and nourishes the cartilage and helps to reduce friction within the joint during movement.

The synovial membrane has a highly specialized structure, consisting of two layers: the intima and the subintima. The intima is a thin layer of cells that are in direct contact with the synovial fluid, while the subintima is a more fibrous layer that contains blood vessels and nerves.

The main function of the synovial membrane is to produce and regulate the production of synovial fluid, as well as to provide nutrients to the articular cartilage. It also plays a role in the immune response within the joint, helping to protect against infection and inflammation. However, abnormalities in the synovial membrane can lead to conditions such as rheumatoid arthritis, where the membrane becomes inflamed and produces excess synovial fluid, leading to pain, swelling, and joint damage.

Microbial viability is the ability of a microorganism to grow, reproduce and maintain its essential life functions. It can be determined through various methods such as cell growth in culture media, staining techniques that detect metabolic activity, or direct observation of active movement. In contrast, non-viable microorganisms are those that have been killed or inactivated and cannot replicate or cause further harm. The measurement of microbial viability is important in various fields such as medicine, food safety, water quality, and environmental monitoring to assess the effectiveness of disinfection and sterilization procedures, and to determine the presence and concentration of harmful bacteria in different environments.

Neutrophil infiltration is a pathological process characterized by the accumulation of neutrophils, a type of white blood cell, in tissue. It is a common feature of inflammation and occurs in response to infection, injury, or other stimuli that trigger an immune response. Neutrophils are attracted to the site of tissue damage by chemical signals called chemokines, which are released by damaged cells and activated immune cells. Once they reach the site of inflammation, neutrophils help to clear away damaged tissue and microorganisms through a process called phagocytosis. However, excessive or prolonged neutrophil infiltration can also contribute to tissue damage and may be associated with various disease states, including cancer, autoimmune disorders, and ischemia-reperfusion injury.

Bronchoalveolar lavage (BAL) is a medical procedure in which a small amount of fluid is introduced into a segment of the lung and then gently suctioned back out. The fluid contains cells and other materials that can be analyzed to help diagnose various lung conditions, such as inflammation, infection, or cancer.

The procedure is typically performed during bronchoscopy, which involves inserting a thin, flexible tube with a light and camera on the end through the nose or mouth and into the lungs. Once the bronchoscope is in place, a small catheter is passed through the bronchoscope and into the desired lung segment. The fluid is then introduced and suctioned back out, and the sample is sent to a laboratory for analysis.

BAL can be helpful in diagnosing various conditions such as pneumonia, interstitial lung diseases, alveolar proteinosis, and some types of cancer. It can also be used to monitor the effectiveness of treatment for certain lung conditions. However, like any medical procedure, it carries some risks, including bleeding, infection, and respiratory distress. Therefore, it is important that the procedure is performed by a qualified healthcare professional in a controlled setting.

Interferon-beta (IFN-β) is a type of cytokine - specifically, it's a protein that is produced and released by cells in response to stimulation by a virus or other foreign substance. It belongs to the interferon family of cytokines, which play important roles in the body's immune response to infection.

IFN-β has antiviral properties and helps to regulate the immune system. It works by binding to specific receptors on the surface of cells, which triggers a signaling cascade that leads to the activation of genes involved in the antiviral response. This results in the production of proteins that inhibit viral replication and promote the death of infected cells.

IFN-β is used as a medication for the treatment of certain autoimmune diseases, such as multiple sclerosis (MS). In MS, the immune system mistakenly attacks the protective coating around nerve fibers in the brain and spinal cord, causing inflammation and damage to the nerves. IFN-β has been shown to reduce the frequency and severity of relapses in people with MS, possibly by modulating the immune response and reducing inflammation.

It's important to note that while IFN-β is an important component of the body's natural defense system, it can also have side effects when used as a medication. Common side effects of IFN-β therapy include flu-like symptoms such as fever, chills, and muscle aches, as well as injection site reactions. More serious side effects are rare but can occur, so it's important to discuss the risks and benefits of this treatment with a healthcare provider.

Disease susceptibility, also known as genetic predisposition or genetic susceptibility, refers to the increased likelihood or risk of developing a particular disease due to inheriting specific genetic variations or mutations. These genetic factors can make an individual more vulnerable to certain diseases compared to those who do not have these genetic changes.

It is important to note that having a genetic predisposition does not guarantee that a person will definitely develop the disease. Other factors, such as environmental exposures, lifestyle choices, and additional genetic variations, can influence whether or not the disease will manifest. In some cases, early detection and intervention may help reduce the risk or delay the onset of the disease in individuals with a known genetic susceptibility.

Experimental neoplasms refer to abnormal growths or tumors that are induced and studied in a controlled laboratory setting, typically in animals or cell cultures. These studies are conducted to understand the fundamental mechanisms of cancer development, progression, and potential treatment strategies. By manipulating various factors such as genetic mutations, environmental exposures, and pharmacological interventions, researchers can gain valuable insights into the complex processes underlying neoplasm formation and identify novel targets for cancer therapy. It is important to note that experimental neoplasms may not always accurately represent human cancers, and further research is needed to translate these findings into clinically relevant applications.

Biological transport refers to the movement of molecules, ions, or solutes across biological membranes or through cells in living organisms. This process is essential for maintaining homeostasis, regulating cellular functions, and enabling communication between cells. There are two main types of biological transport: passive transport and active transport.

Passive transport does not require the input of energy and includes:

1. Diffusion: The random movement of molecules from an area of high concentration to an area of low concentration until equilibrium is reached.
2. Osmosis: The diffusion of solvent molecules (usually water) across a semi-permeable membrane from an area of lower solute concentration to an area of higher solute concentration.
3. Facilitated diffusion: The assisted passage of polar or charged substances through protein channels or carriers in the cell membrane, which increases the rate of diffusion without consuming energy.

Active transport requires the input of energy (in the form of ATP) and includes:

1. Primary active transport: The direct use of ATP to move molecules against their concentration gradient, often driven by specific transport proteins called pumps.
2. Secondary active transport: The coupling of the movement of one substance down its electrochemical gradient with the uphill transport of another substance, mediated by a shared transport protein. This process is also known as co-transport or counter-transport.

"Toxoplasma" is a genus of protozoan parasites, and the most well-known species is "Toxoplasma gondii." This particular species is capable of infecting virtually all warm-blooded animals, including humans. It's known for its complex life cycle that involves felines (cats) as the definitive host.

Infection in humans, called toxoplasmosis, often occurs through ingestion of contaminated food or water, or through contact with cat feces that contain T. gondii oocysts. While many people infected with Toxoplasma show no symptoms, it can cause serious health problems in immunocompromised individuals and developing fetuses if a woman becomes infected during pregnancy.

It's important to note that while I strive to provide accurate information, this definition should not be used for self-diagnosis or treatment. Always consult with a healthcare professional for medical advice.

"Intraperitoneal injection" is a medical term that refers to the administration of a substance or medication directly into the peritoneal cavity, which is the space between the lining of the abdominal wall and the organs contained within it. This type of injection is typically used in clinical settings for various purposes, such as delivering chemotherapy drugs, anesthetics, or other medications directly to the abdominal organs.

The procedure involves inserting a needle through the abdominal wall and into the peritoneal cavity, taking care to avoid any vital structures such as blood vessels or nerves. Once the needle is properly positioned, the medication can be injected slowly and carefully to ensure even distribution throughout the cavity.

It's important to note that intraperitoneal injections are typically reserved for situations where other routes of administration are not feasible or effective, as they carry a higher risk of complications such as infection, bleeding, or injury to surrounding organs. As with any medical procedure, it should only be performed by trained healthcare professionals under appropriate clinical circumstances.

Signal Transducer and Activator of Transcription 1 (STAT1) is a transcription factor that plays a crucial role in the regulation of gene expression in response to cytokines and interferons. It is activated through phosphorylation by Janus kinases (JAKs) upon binding of cytokines to their respective receptors. Once activated, STAT1 forms homodimers or heterodimers with other STAT family members, translocates to the nucleus, and binds to specific DNA sequences called gamma-activated sites (GAS) in the promoter regions of target genes. This results in the modulation of gene expression involved in various cellular processes such as immune responses, differentiation, apoptosis, and cell cycle control. STAT1 also plays a critical role in the antiviral response by mediating the transcription of interferon-stimulated genes (ISGs).

'Tumor cells, cultured' refers to the process of removing cancerous cells from a tumor and growing them in controlled laboratory conditions. This is typically done by isolating the tumor cells from a patient's tissue sample, then placing them in a nutrient-rich environment that promotes their growth and multiplication.

The resulting cultured tumor cells can be used for various research purposes, including the study of cancer biology, drug development, and toxicity testing. They provide a valuable tool for researchers to better understand the behavior and characteristics of cancer cells outside of the human body, which can lead to the development of more effective cancer treatments.

It is important to note that cultured tumor cells may not always behave exactly the same way as they do in the human body, so findings from cell culture studies must be validated through further research, such as animal models or clinical trials.

A ligand, in the context of biochemistry and medicine, is a molecule that binds to a specific site on a protein or a larger biomolecule, such as an enzyme or a receptor. This binding interaction can modify the function or activity of the target protein, either activating it or inhibiting it. Ligands can be small molecules, like hormones or neurotransmitters, or larger structures, like antibodies. The study of ligand-protein interactions is crucial for understanding cellular processes and developing drugs, as many therapeutic compounds function by binding to specific targets within the body.

Peritonitis is a medical condition characterized by inflammation of the peritoneum, which is the serous membrane that lines the inner wall of the abdominal cavity and covers the abdominal organs. The peritoneum has an important role in protecting the abdominal organs and providing a smooth surface for them to move against each other.

Peritonitis can occur as a result of bacterial or fungal infection, chemical irritation, or trauma to the abdomen. The most common cause of peritonitis is a rupture or perforation of an organ in the abdominal cavity, such as the appendix, stomach, or intestines, which allows bacteria from the gut to enter the peritoneal cavity.

Symptoms of peritonitis may include abdominal pain and tenderness, fever, nausea and vomiting, loss of appetite, and decreased bowel movements. In severe cases, peritonitis can lead to sepsis, a life-threatening condition characterized by widespread inflammation throughout the body.

Treatment for peritonitis typically involves antibiotics to treat the infection, as well as surgical intervention to repair any damage to the abdominal organs and remove any infected fluid or tissue from the peritoneal cavity. In some cases, a temporary or permanent drain may be placed in the abdomen to help remove excess fluid and promote healing.

Transforming Growth Factor-beta (TGF-β) is a type of cytokine, which is a cell signaling protein involved in the regulation of various cellular processes, including cell growth, differentiation, and apoptosis (programmed cell death). TGF-β plays a critical role in embryonic development, tissue homeostasis, and wound healing. It also has been implicated in several pathological conditions such as fibrosis, cancer, and autoimmune diseases.

TGF-β exists in multiple isoforms (TGF-β1, TGF-β2, and TGF-β3) that are produced by many different cell types, including immune cells, epithelial cells, and fibroblasts. The protein is synthesized as a precursor molecule, which is cleaved to release the active TGF-β peptide. Once activated, TGF-β binds to its receptors on the cell surface, leading to the activation of intracellular signaling pathways that regulate gene expression and cell behavior.

In summary, Transforming Growth Factor-beta (TGF-β) is a multifunctional cytokine involved in various cellular processes, including cell growth, differentiation, apoptosis, embryonic development, tissue homeostasis, and wound healing. It has been implicated in several pathological conditions such as fibrosis, cancer, and autoimmune diseases.

Real-Time Polymerase Chain Reaction (RT-PCR) is a laboratory technique used in molecular biology to amplify and detect specific DNA sequences in real-time. It is a sensitive and specific method that allows for the quantification of target nucleic acids, such as DNA or RNA, through the use of fluorescent reporter molecules.

The RT-PCR process involves several steps: first, the template DNA is denatured to separate the double-stranded DNA into single strands. Then, primers (short sequences of DNA) specific to the target sequence are added and allowed to anneal to the template DNA. Next, a heat-stable enzyme called Taq polymerase adds nucleotides to the annealed primers, extending them along the template DNA until a new double-stranded DNA molecule is formed.

During each amplification cycle, fluorescent reporter molecules are added that bind specifically to the newly synthesized DNA. As more and more copies of the target sequence are generated, the amount of fluorescence increases in proportion to the number of copies present. This allows for real-time monitoring of the PCR reaction and quantification of the target nucleic acid.

RT-PCR is commonly used in medical diagnostics, research, and forensics to detect and quantify specific DNA or RNA sequences. It has been widely used in the diagnosis of infectious diseases, genetic disorders, and cancer, as well as in the identification of microbial pathogens and the detection of gene expression.

Tuberculosis (TB) is a chronic infectious disease caused by the bacterium Mycobacterium tuberculosis. It primarily affects the lungs but can also involve other organs and tissues in the body. The infection is usually spread through the air when an infected person coughs, sneezes, or talks.

The symptoms of pulmonary TB include persistent cough, chest pain, coughing up blood, fatigue, fever, night sweats, and weight loss. Diagnosis typically involves a combination of medical history, physical examination, chest X-ray, and microbiological tests such as sputum smear microscopy and culture. In some cases, molecular tests like polymerase chain reaction (PCR) may be used for rapid diagnosis.

Treatment usually consists of a standard six-month course of multiple antibiotics, including isoniazid, rifampin, ethambutol, and pyrazinamide. In some cases, longer treatment durations or different drug regimens might be necessary due to drug resistance or other factors. Preventive measures include vaccination with the Bacillus Calmette-Guérin (BCG) vaccine and early detection and treatment of infected individuals to prevent transmission.

Concanavalin A (Con A) is a type of protein known as a lectin, which is found in the seeds of the plant Canavalia ensiformis, also known as jack bean. It is often used in laboratory settings as a tool to study various biological processes, such as cell division and the immune response, due to its ability to bind specifically to certain sugars on the surface of cells. Con A has been extensively studied for its potential applications in medicine, including as a possible treatment for cancer and viral infections. However, more research is needed before these potential uses can be realized.

B-lymphocytes, also known as B-cells, are a type of white blood cell that plays a key role in the immune system's response to infection. They are responsible for producing antibodies, which are proteins that help to neutralize or destroy pathogens such as bacteria and viruses.

When a B-lymphocyte encounters a pathogen, it becomes activated and begins to divide and differentiate into plasma cells, which produce and secrete large amounts of antibodies specific to the antigens on the surface of the pathogen. These antibodies bind to the pathogen, marking it for destruction by other immune cells such as neutrophils and macrophages.

B-lymphocytes also have a role in presenting antigens to T-lymphocytes, another type of white blood cell involved in the immune response. This helps to stimulate the activation and proliferation of T-lymphocytes, which can then go on to destroy infected cells or help to coordinate the overall immune response.

Overall, B-lymphocytes are an essential part of the adaptive immune system, providing long-lasting immunity to previously encountered pathogens and helping to protect against future infections.

Polymerase Chain Reaction (PCR) is a laboratory technique used to amplify specific regions of DNA. It enables the production of thousands to millions of copies of a particular DNA sequence in a rapid and efficient manner, making it an essential tool in various fields such as molecular biology, medical diagnostics, forensic science, and research.

The PCR process involves repeated cycles of heating and cooling to separate the DNA strands, allow primers (short sequences of single-stranded DNA) to attach to the target regions, and extend these primers using an enzyme called Taq polymerase, resulting in the exponential amplification of the desired DNA segment.

In a medical context, PCR is often used for detecting and quantifying specific pathogens (viruses, bacteria, fungi, or parasites) in clinical samples, identifying genetic mutations or polymorphisms associated with diseases, monitoring disease progression, and evaluating treatment effectiveness.

Species specificity is a term used in the field of biology, including medicine, to refer to the characteristic of a biological entity (such as a virus, bacterium, or other microorganism) that allows it to interact exclusively or preferentially with a particular species. This means that the biological entity has a strong affinity for, or is only able to infect, a specific host species.

For example, HIV is specifically adapted to infect human cells and does not typically infect other animal species. Similarly, some bacterial toxins are species-specific and can only affect certain types of animals or humans. This concept is important in understanding the transmission dynamics and host range of various pathogens, as well as in developing targeted therapies and vaccines.

Prostaglandin-Endoperoxide Synthases (PTGS), also known as Cyclooxygenases (COX), are a group of enzymes that catalyze the conversion of arachidonic acid into prostaglandin G2 and H2, which are further metabolized to produce various prostaglandins and thromboxanes. These lipid mediators play crucial roles in several physiological processes such as inflammation, pain, fever, and blood clotting. There are two major isoforms of PTGS: PTGS-1 (COX-1) and PTGS-2 (COX-2). While COX-1 is constitutively expressed in most tissues and involved in homeostatic functions, COX-2 is usually induced during inflammation and tissue injury. Nonsteroidal anti-inflammatory drugs (NSAIDs) exert their therapeutic effects by inhibiting these enzymes, thereby reducing the production of prostaglandins and thromboxanes.

DNA-binding proteins are a type of protein that have the ability to bind to DNA (deoxyribonucleic acid), the genetic material of organisms. These proteins play crucial roles in various biological processes, such as regulation of gene expression, DNA replication, repair and recombination.

The binding of DNA-binding proteins to specific DNA sequences is mediated by non-covalent interactions, including electrostatic, hydrogen bonding, and van der Waals forces. The specificity of binding is determined by the recognition of particular nucleotide sequences or structural features of the DNA molecule.

DNA-binding proteins can be classified into several categories based on their structure and function, such as transcription factors, histones, and restriction enzymes. Transcription factors are a major class of DNA-binding proteins that regulate gene expression by binding to specific DNA sequences in the promoter region of genes and recruiting other proteins to modulate transcription. Histones are DNA-binding proteins that package DNA into nucleosomes, the basic unit of chromatin structure. Restriction enzymes are DNA-binding proteins that recognize and cleave specific DNA sequences, and are widely used in molecular biology research and biotechnology applications.

Northern blotting is a laboratory technique used in molecular biology to detect and analyze specific RNA molecules (such as mRNA) in a mixture of total RNA extracted from cells or tissues. This technique is called "Northern" blotting because it is analogous to the Southern blotting method, which is used for DNA detection.

The Northern blotting procedure involves several steps:

1. Electrophoresis: The total RNA mixture is first separated based on size by running it through an agarose gel using electrical current. This separates the RNA molecules according to their length, with smaller RNA fragments migrating faster than larger ones.

2. Transfer: After electrophoresis, the RNA bands are denatured (made single-stranded) and transferred from the gel onto a nitrocellulose or nylon membrane using a technique called capillary transfer or vacuum blotting. This step ensures that the order and relative positions of the RNA fragments are preserved on the membrane, similar to how they appear in the gel.

3. Cross-linking: The RNA is then chemically cross-linked to the membrane using UV light or heat treatment, which helps to immobilize the RNA onto the membrane and prevent it from washing off during subsequent steps.

4. Prehybridization: Before adding the labeled probe, the membrane is prehybridized in a solution containing blocking agents (such as salmon sperm DNA or yeast tRNA) to minimize non-specific binding of the probe to the membrane.

5. Hybridization: A labeled nucleic acid probe, specific to the RNA of interest, is added to the prehybridization solution and allowed to hybridize (form base pairs) with its complementary RNA sequence on the membrane. The probe can be either a DNA or an RNA molecule, and it is typically labeled with a radioactive isotope (such as ³²P) or a non-radioactive label (such as digoxigenin).

6. Washing: After hybridization, the membrane is washed to remove unbound probe and reduce background noise. The washing conditions (temperature, salt concentration, and detergent concentration) are optimized based on the stringency required for specific hybridization.

7. Detection: The presence of the labeled probe is then detected using an appropriate method, depending on the type of label used. For radioactive probes, this typically involves exposing the membrane to X-ray film or a phosphorimager screen and analyzing the resulting image. For non-radioactive probes, detection can be performed using colorimetric, chemiluminescent, or fluorescent methods.

8. Data analysis: The intensity of the signal is quantified and compared to controls (such as housekeeping genes) to determine the relative expression level of the RNA of interest. This information can be used for various purposes, such as identifying differentially expressed genes in response to a specific treatment or comparing gene expression levels across different samples or conditions.

"Wistar rats" are a strain of albino rats that are widely used in laboratory research. They were developed at the Wistar Institute in Philadelphia, USA, and were first introduced in 1906. Wistar rats are outbred, which means that they are genetically diverse and do not have a fixed set of genetic characteristics like inbred strains.

Wistar rats are commonly used as animal models in biomedical research because of their size, ease of handling, and relatively low cost. They are used in a wide range of research areas, including toxicology, pharmacology, nutrition, cancer, cardiovascular disease, and behavioral studies. Wistar rats are also used in safety testing of drugs, medical devices, and other products.

Wistar rats are typically larger than many other rat strains, with males weighing between 500-700 grams and females weighing between 250-350 grams. They have a lifespan of approximately 2-3 years. Wistar rats are also known for their docile and friendly nature, making them easy to handle and work with in the laboratory setting.

Bacterial antigens are substances found on the surface or produced by bacteria that can stimulate an immune response in a host organism. These antigens can be proteins, polysaccharides, teichoic acids, lipopolysaccharides, or other molecules that are recognized as foreign by the host's immune system.

When a bacterial antigen is encountered by the host's immune system, it triggers a series of responses aimed at eliminating the bacteria and preventing infection. The host's immune system recognizes the antigen as foreign through the use of specialized receptors called pattern recognition receptors (PRRs), which are found on various immune cells such as macrophages, dendritic cells, and neutrophils.

Once a bacterial antigen is recognized by the host's immune system, it can stimulate both the innate and adaptive immune responses. The innate immune response involves the activation of inflammatory pathways, the recruitment of immune cells to the site of infection, and the production of antimicrobial peptides.

The adaptive immune response, on the other hand, involves the activation of T cells and B cells, which are specific to the bacterial antigen. These cells can recognize and remember the antigen, allowing for a more rapid and effective response upon subsequent exposures.

Bacterial antigens are important in the development of vaccines, as they can be used to stimulate an immune response without causing disease. By identifying specific bacterial antigens that are associated with virulence or pathogenicity, researchers can develop vaccines that target these antigens and provide protection against infection.

Pneumonia is an infection or inflammation of the alveoli (tiny air sacs) in one or both lungs. It's often caused by bacteria, viruses, or fungi. Accumulated pus and fluid in these air sacs make it difficult to breathe, which can lead to coughing, chest pain, fever, and difficulty breathing. The severity of symptoms can vary from mild to life-threatening, depending on the underlying cause, the patient's overall health, and age. Pneumonia is typically diagnosed through a combination of physical examination, medical history, and diagnostic tests such as chest X-rays or blood tests. Treatment usually involves antibiotics for bacterial pneumonia, antivirals for viral pneumonia, and supportive care like oxygen therapy, hydration, and rest.

Galectin-3 is a type of protein belonging to the galectin family, which binds to carbohydrates (sugars) and plays a role in various biological processes such as inflammation, immune response, and cancer. It is also known as Mac-2 binding protein or LGALS3.

Galectin-3 is unique among galectins because it can form oligomers (complexes of multiple subunits) and has a wide range of functions in the body. It is involved in cell adhesion, proliferation, differentiation, apoptosis (programmed cell death), and angiogenesis (formation of new blood vessels).

In the context of disease, Galectin-3 has been implicated in several pathological conditions such as fibrosis, heart failure, and cancer. High levels of Galectin-3 have been associated with poor prognosis in patients with heart failure, and it is considered a potential biomarker for this condition. In addition, Galectin-3 has been shown to promote tumor growth, angiogenesis, and metastasis, making it a target for cancer therapy.

A cell line that is derived from tumor cells and has been adapted to grow in culture. These cell lines are often used in research to study the characteristics of cancer cells, including their growth patterns, genetic changes, and responses to various treatments. They can be established from many different types of tumors, such as carcinomas, sarcomas, and leukemias. Once established, these cell lines can be grown and maintained indefinitely in the laboratory, allowing researchers to conduct experiments and studies that would not be feasible using primary tumor cells. It is important to note that tumor cell lines may not always accurately represent the behavior of the original tumor, as they can undergo genetic changes during their time in culture.

Sterol O-Acyltransferase (SOAT, also known as ACAT for Acyl-CoA:cholesterol acyltransferase) is an enzyme that plays a crucial role in cholesterol homeostasis within cells. Specifically, it catalyzes the reaction of esterifying free cholesterol with fatty acyl-coenzyme A (fatty acyl-CoA) to form cholesteryl esters. This enzymatic activity allows for the intracellular storage of excess cholesterol in lipid droplets, reducing the levels of free cholesterol in the cell and thus preventing its potential toxic effects on membranes and proteins. There are two isoforms of SOAT, SOAT1 and SOAT2, which exhibit distinct subcellular localization and functions. Dysregulation of SOAT activity has been implicated in various pathological conditions, including atherosclerosis and neurodegenerative disorders.

Osteoclasts are large, multinucleated cells that are primarily responsible for bone resorption, a process in which they break down and dissolve the mineralized matrix of bones. They are derived from monocyte-macrophage precursor cells of hematopoietic origin and play a crucial role in maintaining bone homeostasis by balancing bone formation and bone resorption.

Osteoclasts adhere to the bone surface and create an isolated microenvironment, called the "resorption lacuna," between their cell membrane and the bone surface. Here, they release hydrogen ions into the lacuna through a process called proton pumping, which lowers the pH and dissolves the mineral component of the bone matrix. Additionally, osteoclasts secrete proteolytic enzymes, such as cathepsin K, that degrade the organic components, like collagen, in the bone matrix.

An imbalance in osteoclast activity can lead to various bone diseases, including osteoporosis and Paget's disease, where excessive bone resorption results in weakened and fragile bones.

Arachidonic acid is a type of polyunsaturated fatty acid that is found naturally in the body and in certain foods. It is an essential fatty acid, meaning that it cannot be produced by the human body and must be obtained through the diet. Arachidonic acid is a key component of cell membranes and plays a role in various physiological processes, including inflammation and blood clotting.

In the body, arachidonic acid is released from cell membranes in response to various stimuli, such as injury or infection. Once released, it can be converted into a variety of bioactive compounds, including prostaglandins, thromboxanes, and leukotrienes, which mediate various physiological responses, including inflammation, pain, fever, and blood clotting.

Arachidonic acid is found in high concentrations in animal products such as meat, poultry, fish, and eggs, as well as in some plant sources such as certain nuts and seeds. It is also available as a dietary supplement. However, it is important to note that excessive intake of arachidonic acid can contribute to the development of inflammation and other health problems, so it is recommended to consume this fatty acid in moderation as part of a balanced diet.

Interleukin-13 (IL-13) is a cytokine that plays a crucial role in the immune response, particularly in the development of allergic inflammation and hypersensitivity reactions. It is primarily produced by activated Th2 cells, mast cells, basophils, and eosinophils. IL-13 mediates its effects through binding to the IL-13 receptor complex, which consists of the IL-13Rα1 and IL-4Rα chains.

IL-13 has several functions in the body, including:

* Regulation of IgE production by B cells
* Induction of eosinophil differentiation and activation
* Inhibition of proinflammatory cytokine production by macrophages
* Promotion of mucus production and airway hyperresponsiveness in the lungs, contributing to the pathogenesis of asthma.

Dysregulation of IL-13 has been implicated in various diseases, such as allergic asthma, atopic dermatitis, and chronic rhinosinusitis. Therefore, targeting IL-13 with biologic therapies has emerged as a promising approach for the treatment of these conditions.

Vacuoles are membrane-bound organelles found in the cells of most eukaryotic organisms. They are essentially fluid-filled sacs that store various substances, such as enzymes, waste products, and nutrients. In plants, vacuoles often contain water, ions, and various organic compounds, while in fungi, they may store lipids or pigments. Vacuoles can also play a role in maintaining the turgor pressure of cells, which is critical for cell shape and function.

In animal cells, vacuoles are typically smaller and less numerous than in plant cells. Animal cells have lysosomes, which are membrane-bound organelles that contain digestive enzymes and break down waste materials, cellular debris, and foreign substances. Lysosomes can be considered a type of vacuole, but they are more specialized in their function.

Overall, vacuoles are essential for maintaining the health and functioning of cells by providing a means to store and dispose of various substances.

Interleukin-18 (IL-18) is a pro-inflammatory cytokine, a type of signaling molecule used in intercellular communication. It belongs to the interleukin-1 (IL-1) family and is primarily produced by macrophages, although other cells such as keratinocytes, osteoblasts, and Kupffer cells can also produce it.

IL-18 plays a crucial role in the innate and adaptive immune responses. It contributes to the differentiation of Th1 (T helper 1) cells, which are critical for fighting intracellular pathogens, and enhances the cytotoxic activity of natural killer (NK) cells and CD8+ T cells. IL-18 also has a role in the production of interferon-gamma (IFN-γ), a cytokine that activates immune cells and has antiviral properties.

Dysregulation of IL-18 has been implicated in several inflammatory diseases, such as rheumatoid arthritis, Crohn's disease, and psoriasis. It is also involved in the pathogenesis of some autoimmune disorders and has been investigated as a potential therapeutic target for these conditions.

The aorta is the largest artery in the human body, which originates from the left ventricle of the heart and carries oxygenated blood to the rest of the body. It can be divided into several parts, including the ascending aorta, aortic arch, and descending aorta. The ascending aorta gives rise to the coronary arteries that supply blood to the heart muscle. The aortic arch gives rise to the brachiocephalic, left common carotid, and left subclavian arteries, which supply blood to the head, neck, and upper extremities. The descending aorta travels through the thorax and abdomen, giving rise to various intercostal, visceral, and renal arteries that supply blood to the chest wall, organs, and kidneys.

Mitogen-Activated Protein Kinases (MAPKs) are a family of serine/threonine protein kinases that play crucial roles in various cellular processes, including proliferation, differentiation, transformation, and apoptosis, in response to diverse stimuli such as mitogens, growth factors, hormones, cytokines, and environmental stresses. They are highly conserved across eukaryotes and consist of a three-tiered kinase module composed of MAPK kinase kinases (MAP3Ks), MAPK kinases (MKKs or MAP2Ks), and MAPKs.

Activation of MAPKs occurs through a sequential phosphorylation and activation cascade, where MAP3Ks phosphorylate and activate MKKs, which in turn phosphorylate and activate MAPKs at specific residues (Thr-X-Tyr or Ser-Pro motifs). Once activated, MAPKs can further phosphorylate and regulate various downstream targets, including transcription factors and other protein kinases.

There are four major groups of MAPKs in mammals: extracellular signal-regulated kinases (ERK1/2), c-Jun N-terminal kinases (JNK1/2/3), p38 MAPKs (p38α/β/γ/δ), and ERK5/BMK1. Each group of MAPKs has distinct upstream activators, downstream targets, and cellular functions, allowing for a high degree of specificity in signal transduction and cellular responses. Dysregulation of MAPK signaling pathways has been implicated in various human diseases, including cancer, diabetes, neurodegenerative disorders, and inflammatory diseases.

Intracellular fluid (ICF) refers to the fluid that is contained within the cells of the body. It makes up about two-thirds of the total body water and is found in the cytosol, which is the liquid inside the cell's membrane. The intracellular fluid contains various ions, nutrients, waste products, and other molecules that are necessary for the proper functioning of the cell.

The main ions present in the ICF include potassium (K+), magnesium (Mg2+), and phosphate (HPO42-). The concentration of these ions inside the cell is different from their concentration outside the cell, which creates an electrochemical gradient that plays a crucial role in various physiological processes such as nerve impulse transmission, muscle contraction, and cell volume regulation.

Maintaining the balance of intracellular fluid is essential for normal cell function, and any disruption in this balance can lead to various health issues. Factors that can affect the ICF balance include changes in hydration status, electrolyte imbalances, and certain medical conditions such as kidney disease or heart failure.

A leukocyte count, also known as a white blood cell (WBC) count, is a laboratory test that measures the number of leukocytes in a sample of blood. Leukocytes are a vital part of the body's immune system and help fight infection and inflammation. A high or low leukocyte count may indicate an underlying medical condition, such as an infection, inflammation, or a bone marrow disorder. The normal range for a leukocyte count in adults is typically between 4,500 and 11,000 cells per microliter (mcL) of blood. However, the normal range can vary slightly depending on the laboratory and the individual's age and sex.

Immune tolerance, also known as immunological tolerance or specific immune tolerance, is a state of unresponsiveness or non-reactivity of the immune system towards a particular substance (antigen) that has the potential to elicit an immune response. This occurs when the immune system learns to distinguish "self" from "non-self" and does not attack the body's own cells, tissues, and organs.

In the context of transplantation, immune tolerance refers to the absence of a destructive immune response towards the transplanted organ or tissue, allowing for long-term graft survival without the need for immunosuppressive therapy. Immune tolerance can be achieved through various strategies, including hematopoietic stem cell transplantation, costimulation blockade, and regulatory T cell induction.

In summary, immune tolerance is a critical mechanism that prevents the immune system from attacking the body's own structures while maintaining the ability to respond appropriately to foreign pathogens and antigens.

An antigen is a substance (usually a protein) that is recognized as foreign by the immune system and stimulates an immune response, leading to the production of antibodies or activation of T-cells. Antigens can be derived from various sources, including bacteria, viruses, fungi, parasites, and tumor cells. They can also come from non-living substances such as pollen, dust mites, or chemicals.

Antigens contain epitopes, which are specific regions on the antigen molecule that are recognized by the immune system. The immune system's response to an antigen depends on several factors, including the type of antigen, its size, and its location in the body.

In general, antigens can be classified into two main categories:

1. T-dependent antigens: These require the help of T-cells to stimulate an immune response. They are typically larger, more complex molecules that contain multiple epitopes capable of binding to both MHC class II molecules on antigen-presenting cells and T-cell receptors on CD4+ T-cells.
2. T-independent antigens: These do not require the help of T-cells to stimulate an immune response. They are usually smaller, simpler molecules that contain repetitive epitopes capable of cross-linking B-cell receptors and activating them directly.

Understanding antigens and their properties is crucial for developing vaccines, diagnostic tests, and immunotherapies.

"Francisella tularensis" is a gram-negative, aerobic, coccobacillus bacterium that is the etiological agent of tularemia. It is highly infectious and can be transmitted to humans through various routes such as contact with infected animals, ingestion of contaminated food or water, inhalation of contaminated aerosols, or bites from infected arthropods. The bacterium can cause a range of clinical manifestations depending on the route of infection and includes ulceroglandular, oculoglandular, oropharyngeal, pneumonic, and typhoidal tularemia. "Francisella tularensis" is considered a potential bioterrorism agent due to its high infectivity and potential for causing severe illness and death.

Cathepsins are a type of proteolytic enzymes, which are found in lysosomes and are responsible for breaking down proteins inside the cell. They are classified as papain-like cysteine proteases and play important roles in various physiological processes, including tissue remodeling, antigen presentation, and apoptosis (programmed cell death). There are several different types of cathepsins, including cathepsin B, C, D, F, H, K, L, S, V, and X/Z, each with distinct substrate specificities and functions.

Dysregulation of cathepsins has been implicated in various pathological conditions, such as cancer, neurodegenerative diseases, and inflammatory disorders. For example, overexpression or hyperactivation of certain cathepsins has been shown to contribute to tumor invasion and metastasis, while their inhibition has been explored as a potential therapeutic strategy in cancer treatment. Similarly, abnormal levels of cathepsins have been linked to the progression of neurodegenerative diseases like Alzheimer's and Parkinson's, making them attractive targets for drug development.

Chemokines are a family of small signaling proteins that are involved in immune regulation and inflammation. They mediate their effects by interacting with specific cell surface receptors, leading to the activation and migration of various types of immune cells. Chemokines can be divided into four subfamilies based on the arrangement of conserved cysteine residues near the N-terminus: CXC, CC, C, and CX3C.

CXC chemokines are characterized by the presence of a single amino acid (X) between the first two conserved cysteine residues. They play important roles in the recruitment and activation of neutrophils, which are critical effector cells in the early stages of inflammation. CXC chemokines can be further divided into two subgroups based on the presence or absence of a specific amino acid sequence (ELR motif) near the N-terminus: ELR+ and ELR-.

ELR+ CXC chemokines, such as IL-8, are potent chemoattractants for neutrophils and play important roles in the recruitment of these cells to sites of infection or injury. They bind to and activate the CXCR1 and CXCR2 receptors on the surface of neutrophils, leading to their migration towards the source of the chemokine.

ELR- CXC chemokines, such as IP-10 and MIG, are involved in the recruitment of T cells and other immune cells to sites of inflammation. They bind to and activate different receptors, such as CXCR3, on the surface of these cells, leading to their migration towards the source of the chemokine.

Overall, CXC chemokines play important roles in the regulation of immune responses and inflammation, and dysregulation of their expression or activity has been implicated in a variety of diseases, including cancer, autoimmune disorders, and infectious diseases.

Cricetinae is a subfamily of rodents that includes hamsters, gerbils, and relatives. These small mammals are characterized by having short limbs, compact bodies, and cheek pouches for storing food. They are native to various parts of the world, particularly in Europe, Asia, and Africa. Some species are popular pets due to their small size, easy care, and friendly nature. In a medical context, understanding the biology and behavior of Cricetinae species can be important for individuals who keep them as pets or for researchers studying their physiology.

Immunologic factors refer to the elements of the immune system that contribute to the body's defense against foreign substances, infectious agents, and cancerous cells. These factors include various types of white blood cells (such as lymphocytes, neutrophils, monocytes, and eosinophils), antibodies, complement proteins, cytokines, and other molecules involved in the immune response.

Immunologic factors can be categorized into two main types: innate immunity and adaptive immunity. Innate immunity is the non-specific defense mechanism that provides immediate protection against pathogens through physical barriers (e.g., skin, mucous membranes), chemical barriers (e.g., stomach acid, enzymes), and inflammatory responses. Adaptive immunity, on the other hand, is a specific defense mechanism that develops over time as the immune system learns to recognize and respond to particular pathogens or antigens.

Abnormalities in immunologic factors can lead to various medical conditions, such as autoimmune disorders, immunodeficiency diseases, and allergies. Therefore, understanding immunologic factors is crucial for diagnosing and treating these conditions.

Luminescent measurements refer to the quantitative assessment of the emission of light from a substance that has been excited, typically through some form of energy input such as electrical energy or radiation. In the context of medical diagnostics and research, luminescent measurements can be used in various applications, including bioluminescence imaging, which is used to study biological processes at the cellular and molecular level.

Bioluminescence occurs when a chemical reaction produces light within a living organism, often through the action of enzymes such as luciferase. By introducing a luciferase gene into cells or organisms, researchers can use bioluminescent measurements to track cellular processes and monitor gene expression in real time.

Luminescent measurements may also be used in medical research to study the properties of materials used in medical devices, such as LEDs or optical fibers, or to develop new diagnostic tools based on light-emitting nanoparticles or other luminescent materials.

In summary, luminescent measurements are a valuable tool in medical research and diagnostics, providing a non-invasive way to study biological processes and develop new technologies for disease detection and treatment.

Scanning electron microscopy (SEM) is a type of electron microscopy that uses a focused beam of electrons to scan the surface of a sample and produce a high-resolution image. In SEM, a beam of electrons is scanned across the surface of a specimen, and secondary electrons are emitted from the sample due to interactions between the electrons and the atoms in the sample. These secondary electrons are then detected by a detector and used to create an image of the sample's surface topography. SEM can provide detailed images of the surface of a wide range of materials, including metals, polymers, ceramics, and biological samples. It is commonly used in materials science, biology, and electronics for the examination and analysis of surfaces at the micro- and nanoscale.

Cell culture is a technique used in scientific research to grow and maintain cells from plants, animals, or humans in a controlled environment outside of their original organism. This environment typically consists of a sterile container called a cell culture flask or plate, and a nutrient-rich liquid medium that provides the necessary components for the cells' growth and survival, such as amino acids, vitamins, minerals, and hormones.

There are several different types of cell culture techniques used in research, including:

1. Adherent cell culture: In this technique, cells are grown on a flat surface, such as the bottom of a tissue culture dish or flask. The cells attach to the surface and spread out, forming a monolayer that can be observed and manipulated under a microscope.
2. Suspension cell culture: In suspension culture, cells are grown in liquid medium without any attachment to a solid surface. These cells remain suspended in the medium and can be agitated or mixed to ensure even distribution of nutrients.
3. Organoid culture: Organoids are three-dimensional structures that resemble miniature organs and are grown from stem cells or other progenitor cells. They can be used to study organ development, disease processes, and drug responses.
4. Co-culture: In co-culture, two or more different types of cells are grown together in the same culture dish or flask. This technique is used to study cell-cell interactions and communication.
5. Conditioned medium culture: In this technique, cells are grown in a medium that has been conditioned by previous cultures of other cells. The conditioned medium contains factors secreted by the previous cells that can influence the growth and behavior of the new cells.

Cell culture techniques are widely used in biomedical research to study cellular processes, develop drugs, test toxicity, and investigate disease mechanisms. However, it is important to note that cell cultures may not always accurately represent the behavior of cells in a living organism, and results from cell culture experiments should be validated using other methods.

Growth substances, in the context of medical terminology, typically refer to natural hormones or chemically synthesized agents that play crucial roles in controlling and regulating cell growth, differentiation, and division. They are also known as "growth factors" or "mitogens." These substances include:

1. Proteins: Examples include insulin-like growth factors (IGFs), transforming growth factor-beta (TGF-β), platelet-derived growth factor (PDGF), and fibroblast growth factors (FGFs). They bind to specific receptors on the cell surface, activating intracellular signaling pathways that promote cell proliferation, differentiation, and survival.

2. Steroids: Certain steroid hormones, such as androgens and estrogens, can also act as growth substances by binding to nuclear receptors and influencing gene expression related to cell growth and division.

3. Cytokines: Some cytokines, like interleukins (ILs) and hematopoietic growth factors (HGFs), contribute to the regulation of hematopoiesis, immune responses, and inflammation, thus indirectly affecting cell growth and differentiation.

These growth substances have essential roles in various physiological processes, such as embryonic development, tissue repair, and wound healing. However, abnormal or excessive production or response to these growth substances can lead to pathological conditions, including cancer, benign tumors, and other proliferative disorders.

Interferon type I is a class of signaling proteins, also known as cytokines, that are produced and released by cells in response to the presence of pathogens such as viruses, bacteria, and parasites. These interferons play a crucial role in the body's innate immune system and help to establish an antiviral state in surrounding cells to prevent the spread of infection.

Interferon type I includes several subtypes, such as interferon-alpha (IFN-α), interferon-beta (IFN-β), and interferon-omega (IFN-ω). When produced, these interferons bind to specific receptors on the surface of nearby cells, triggering a cascade of intracellular signaling events that lead to the activation of genes involved in the antiviral response.

The activation of these genes results in the production of enzymes that inhibit viral replication and promote the destruction of infected cells. Interferon type I also enhances the adaptive immune response by promoting the activation and proliferation of immune cells such as T-cells and natural killer (NK) cells, which can directly target and eliminate infected cells.

Overall, interferon type I plays a critical role in the body's defense against viral infections and is an important component of the immune response to many different types of pathogens.

Phosphatidylserines are a type of phospholipids that are essential components of the cell membrane, particularly in the brain. They play a crucial role in maintaining the fluidity and permeability of the cell membrane, and are involved in various cellular processes such as signal transduction, protein anchorage, and apoptosis (programmed cell death). Phosphatidylserines contain a polar head group made up of serine amino acids and two non-polar fatty acid tails. They are abundant in the inner layer of the cell membrane but can be externalized to the outer layer during apoptosis, where they serve as signals for recognition and removal of dying cells by the immune system. Phosphatidylserines have been studied for their potential benefits in various medical conditions, including cognitive decline, Alzheimer's disease, and depression.

PPAR gamma, or Peroxisome Proliferator-Activated Receptor gamma, is a nuclear receptor protein that functions as a transcription factor. It plays a crucial role in the regulation of genes involved in adipogenesis (the process of forming mature fat cells), lipid metabolism, insulin sensitivity, and glucose homeostasis. PPAR gamma is primarily expressed in adipose tissue but can also be found in other tissues such as the immune system, large intestine, and brain.

PPAR gamma forms a heterodimer with another nuclear receptor protein, RXR (Retinoid X Receptor), and binds to specific DNA sequences called PPREs (Peroxisome Proliferator Response Elements) in the promoter regions of target genes. Upon binding, PPAR gamma modulates the transcription of these genes, either activating or repressing their expression.

Agonists of PPAR gamma, such as thiazolidinediones (TZDs), are used clinically to treat type 2 diabetes due to their insulin-sensitizing effects. These drugs work by binding to and activating PPAR gamma, which in turn leads to the upregulation of genes involved in glucose uptake and metabolism in adipose tissue and skeletal muscle.

In summary, PPAR gamma is a nuclear receptor protein that regulates gene expression related to adipogenesis, lipid metabolism, insulin sensitivity, and glucose homeostasis. Its activation has therapeutic implications for the treatment of type 2 diabetes and other metabolic disorders.

Pulmonary fibrosis is a specific type of lung disease that results from the thickening and scarring of the lung tissues, particularly those in the alveoli (air sacs) and interstitium (the space around the air sacs). This scarring makes it harder for the lungs to properly expand and transfer oxygen into the bloodstream, leading to symptoms such as shortness of breath, coughing, fatigue, and eventually respiratory failure. The exact cause of pulmonary fibrosis can vary, with some cases being idiopathic (without a known cause) or related to environmental factors, medications, medical conditions, or genetic predisposition.

In medical terms, the skin is the largest organ of the human body. It consists of two main layers: the epidermis (outer layer) and dermis (inner layer), as well as accessory structures like hair follicles, sweat glands, and oil glands. The skin plays a crucial role in protecting us from external factors such as bacteria, viruses, and environmental hazards, while also regulating body temperature and enabling the sense of touch.

Wound healing is a complex and dynamic process that occurs after tissue injury, aiming to restore the integrity and functionality of the damaged tissue. It involves a series of overlapping phases: hemostasis, inflammation, proliferation, and remodeling.

1. Hemostasis: This initial phase begins immediately after injury and involves the activation of the coagulation cascade to form a clot, which stabilizes the wound and prevents excessive blood loss.
2. Inflammation: Activated inflammatory cells, such as neutrophils and monocytes/macrophages, infiltrate the wound site to eliminate pathogens, remove debris, and release growth factors that promote healing. This phase typically lasts for 2-5 days post-injury.
3. Proliferation: In this phase, various cell types, including fibroblasts, endothelial cells, and keratinocytes, proliferate and migrate to the wound site to synthesize extracellular matrix (ECM) components, form new blood vessels (angiogenesis), and re-epithelialize the wounded area. This phase can last up to several weeks depending on the size and severity of the wound.
4. Remodeling: The final phase of wound healing involves the maturation and realignment of collagen fibers, leading to the restoration of tensile strength in the healed tissue. This process can continue for months to years after injury, although the tissue may never fully regain its original structure and function.

It is important to note that wound healing can be compromised by several factors, including age, nutrition, comorbidities (e.g., diabetes, vascular disease), and infection, which can result in delayed healing or non-healing chronic wounds.

Histochemistry is the branch of pathology that deals with the microscopic localization of cellular or tissue components using specific chemical reactions. It involves the application of chemical techniques to identify and locate specific biomolecules within tissues, cells, and subcellular structures. This is achieved through the use of various staining methods that react with specific antigens or enzymes in the sample, allowing for their visualization under a microscope. Histochemistry is widely used in diagnostic pathology to identify different types of tissues, cells, and structures, as well as in research to study cellular and molecular processes in health and disease.

Culture media is a substance that is used to support the growth of microorganisms or cells in an artificial environment, such as a petri dish or test tube. It typically contains nutrients and other factors that are necessary for the growth and survival of the organisms being cultured. There are many different types of culture media, each with its own specific formulation and intended use. Some common examples include blood agar, which is used to culture bacteria; Sabouraud dextrose agar, which is used to culture fungi; and Eagle's minimum essential medium, which is used to culture animal cells.

Interleukins (ILs) are a group of naturally occurring proteins that are important in the immune system. They are produced by various cells, including immune cells like lymphocytes and macrophages, and they help regulate the immune response by facilitating communication between different types of cells. Interleukins can have both pro-inflammatory and anti-inflammatory effects, depending on the specific interleukin and the context in which it is produced. They play a role in various biological processes, including the development of immune responses, inflammation, and hematopoiesis (the formation of blood cells).

There are many different interleukins that have been identified, and they are numbered according to the order in which they were discovered. For example, IL-1, IL-2, IL-3, etc. Each interleukin has a specific set of functions and targets certain types of cells. Dysregulation of interleukins has been implicated in various diseases, including autoimmune disorders, infections, and cancer.

BCG (Bacillus Calmette-Guérin) vaccine is a type of immunization used primarily to prevent tuberculosis (TB). It contains a live but weakened strain of Mycobacterium bovis, which is related to the bacterium that causes TB in humans (Mycobacterium tuberculosis).

The BCG vaccine works by stimulating an immune response in the body, enabling it to better resist infection with TB bacteria if exposed in the future. It is often given to infants and children in countries where TB is common, and its use varies depending on the national immunization policies. The protection offered by the BCG vaccine is moderate and may not last for a very long time.

In addition to its use against TB, the BCG vaccine has also been investigated for its potential therapeutic role in treating bladder cancer and some other types of cancer. The mechanism of action in these cases is thought to be related to the vaccine's ability to stimulate an immune response against abnormal cells.

Isoenzymes, also known as isoforms, are multiple forms of an enzyme that catalyze the same chemical reaction but differ in their amino acid sequence, structure, and/or kinetic properties. They are encoded by different genes or alternative splicing of the same gene. Isoenzymes can be found in various tissues and organs, and they play a crucial role in biological processes such as metabolism, detoxification, and cell signaling. Measurement of isoenzyme levels in body fluids (such as blood) can provide valuable diagnostic information for certain medical conditions, including tissue damage, inflammation, and various diseases.

An inflammasome is a large cytosolic protein complex that plays a crucial role in the innate immune system's response to infection and stress. It is responsible for the activation of caspase-1, which subsequently leads to the processing and secretion of proinflammatory cytokines, such as interleukin (IL)-1β and IL-18, and the induction of a form of cell death known as pyroptosis.

The inflammasome is formed when certain pattern recognition receptors (PRRs), such as NOD-like receptors (NLRs) or AIM2-like receptors (ALRs), recognize specific pathogen-associated molecular patterns (PAMPs) or damage-associated molecular patterns (DAMPs). This interaction results in the recruitment and assembly of the inflammasome complex, which includes the adaptor protein ASC and pro-caspase-1.

Once activated, caspase-1 cleaves pro-IL-1β and pro-IL-18 into their active forms, which are then released from the cell to recruit immune cells and initiate an inflammatory response. Dysregulation of inflammasome activation has been implicated in various diseases, including autoinflammatory disorders, autoimmune diseases, and neurodegenerative conditions.

Antigen presentation is the process by which certain cells in the immune system, known as antigen presenting cells (APCs), display foreign or abnormal proteins (antigens) on their surface to other immune cells, such as T-cells. This process allows the immune system to recognize and mount a response against harmful pathogens, infected or damaged cells.

There are two main types of antigen presentation: major histocompatibility complex (MHC) class I and MHC class II presentation.

1. MHC class I presentation: APCs, such as dendritic cells, macrophages, and B-cells, process and load antigens onto MHC class I molecules, which are expressed on the surface of almost all nucleated cells in the body. The MHC class I-antigen complex is then recognized by CD8+ T-cells (cytotoxic T-cells), leading to the destruction of infected or damaged cells.
2. MHC class II presentation: APCs, particularly dendritic cells and B-cells, process and load antigens onto MHC class II molecules, which are mainly expressed on the surface of professional APCs. The MHC class II-antigen complex is then recognized by CD4+ T-cells (helper T-cells), leading to the activation of other immune cells, such as B-cells and macrophages, to eliminate the pathogen or damaged cells.

In summary, antigen presentation is a crucial step in the adaptive immune response, allowing for the recognition and elimination of foreign or abnormal substances that could potentially harm the body.

Small interfering RNA (siRNA) is a type of short, double-stranded RNA molecule that plays a role in the RNA interference (RNAi) pathway. The RNAi pathway is a natural cellular process that regulates gene expression by targeting and destroying specific messenger RNA (mRNA) molecules, thereby preventing the translation of those mRNAs into proteins.

SiRNAs are typically 20-25 base pairs in length and are generated from longer double-stranded RNA precursors called hairpin RNAs or dsRNAs by an enzyme called Dicer. Once generated, siRNAs associate with a protein complex called the RNA-induced silencing complex (RISC), which uses one strand of the siRNA (the guide strand) to recognize and bind to complementary sequences in the target mRNA. The RISC then cleaves the target mRNA, leading to its degradation and the inhibition of protein synthesis.

SiRNAs have emerged as a powerful tool for studying gene function and have shown promise as therapeutic agents for a variety of diseases, including viral infections, cancer, and genetic disorders. However, their use as therapeutics is still in the early stages of development, and there are challenges associated with delivering siRNAs to specific cells and tissues in the body.

Biological models, also known as physiological models or organismal models, are simplified representations of biological systems, processes, or mechanisms that are used to understand and explain the underlying principles and relationships. These models can be theoretical (conceptual or mathematical) or physical (such as anatomical models, cell cultures, or animal models). They are widely used in biomedical research to study various phenomena, including disease pathophysiology, drug action, and therapeutic interventions.

Examples of biological models include:

1. Mathematical models: These use mathematical equations and formulas to describe complex biological systems or processes, such as population dynamics, metabolic pathways, or gene regulation networks. They can help predict the behavior of these systems under different conditions and test hypotheses about their underlying mechanisms.
2. Cell cultures: These are collections of cells grown in a controlled environment, typically in a laboratory dish or flask. They can be used to study cellular processes, such as signal transduction, gene expression, or metabolism, and to test the effects of drugs or other treatments on these processes.
3. Animal models: These are living organisms, usually vertebrates like mice, rats, or non-human primates, that are used to study various aspects of human biology and disease. They can provide valuable insights into the pathophysiology of diseases, the mechanisms of drug action, and the safety and efficacy of new therapies.
4. Anatomical models: These are physical representations of biological structures or systems, such as plastic models of organs or tissues, that can be used for educational purposes or to plan surgical procedures. They can also serve as a basis for developing more sophisticated models, such as computer simulations or 3D-printed replicas.

Overall, biological models play a crucial role in advancing our understanding of biology and medicine, helping to identify new targets for therapeutic intervention, develop novel drugs and treatments, and improve human health.

Th1 cells, or Type 1 T helper cells, are a subset of CD4+ T cells that play a crucial role in the cell-mediated immune response. They are characterized by the production of specific cytokines, such as interferon-gamma (IFN-γ), tumor necrosis factor-alpha (TNF-α), and interleukin-2 (IL-2). Th1 cells are essential for protecting against intracellular pathogens, including viruses, bacteria, and parasites. They activate macrophages to destroy ingested microorganisms, stimulate the differentiation of B cells into plasma cells that produce antibodies, and recruit other immune cells to the site of infection. Dysregulation of Th1 cell responses has been implicated in various autoimmune diseases, such as multiple sclerosis, rheumatoid arthritis, and type 1 diabetes.

Transcription factors are proteins that play a crucial role in regulating gene expression by controlling the transcription of DNA to messenger RNA (mRNA). They function by binding to specific DNA sequences, known as response elements, located in the promoter region or enhancer regions of target genes. This binding can either activate or repress the initiation of transcription, depending on the properties and interactions of the particular transcription factor. Transcription factors often act as part of a complex network of regulatory proteins that determine the precise spatiotemporal patterns of gene expression during development, differentiation, and homeostasis in an organism.

A foreign-body reaction is an immune response that occurs when a non-native substance, or "foreign body," is introduced into the human body. This can include things like splinters, surgical implants, or even injected medications. The immune system recognizes these substances as foreign and mounts a response to try to eliminate them.

The initial response to a foreign body is often an acute inflammatory reaction, characterized by the release of chemical mediators that cause vasodilation, increased blood flow, and the migration of white blood cells to the site. This can result in symptoms such as redness, swelling, warmth, and pain.

If the foreign body is not eliminated, a chronic inflammatory response may develop, which can lead to the formation of granulation tissue, fibrosis, and encapsulation of the foreign body. In some cases, this reaction can cause significant tissue damage or impede proper healing.

It's worth noting that not all foreign bodies necessarily elicit a strong immune response. The nature and size of the foreign body, as well as its location in the body, can all influence the severity of the reaction.

Oligonucleotide Array Sequence Analysis is a type of microarray analysis that allows for the simultaneous measurement of the expression levels of thousands of genes in a single sample. In this technique, oligonucleotides (short DNA sequences) are attached to a solid support, such as a glass slide, in a specific pattern. These oligonucleotides are designed to be complementary to specific target mRNA sequences from the sample being analyzed.

During the analysis, labeled RNA or cDNA from the sample is hybridized to the oligonucleotide array. The level of hybridization is then measured and used to determine the relative abundance of each target sequence in the sample. This information can be used to identify differences in gene expression between samples, which can help researchers understand the underlying biological processes involved in various diseases or developmental stages.

It's important to note that this technique requires specialized equipment and bioinformatics tools for data analysis, as well as careful experimental design and validation to ensure accurate and reproducible results.

Proteins are complex, large molecules that play critical roles in the body's functions. They are made up of amino acids, which are organic compounds that are the building blocks of proteins. Proteins are required for the structure, function, and regulation of the body's tissues and organs. They are essential for the growth, repair, and maintenance of body tissues, and they play a crucial role in many biological processes, including metabolism, immune response, and cellular signaling. Proteins can be classified into different types based on their structure and function, such as enzymes, hormones, antibodies, and structural proteins. They are found in various foods, especially animal-derived products like meat, dairy, and eggs, as well as plant-based sources like beans, nuts, and grains.

In the context of medicine, iron is an essential micromineral and key component of various proteins and enzymes. It plays a crucial role in oxygen transport, DNA synthesis, and energy production within the body. Iron exists in two main forms: heme and non-heme. Heme iron is derived from hemoglobin and myoglobin in animal products, while non-heme iron comes from plant sources and supplements.

The recommended daily allowance (RDA) for iron varies depending on age, sex, and life stage:

* For men aged 19-50 years, the RDA is 8 mg/day
* For women aged 19-50 years, the RDA is 18 mg/day
* During pregnancy, the RDA increases to 27 mg/day
* During lactation, the RDA for breastfeeding mothers is 9 mg/day

Iron deficiency can lead to anemia, characterized by fatigue, weakness, and shortness of breath. Excessive iron intake may result in iron overload, causing damage to organs such as the liver and heart. Balanced iron levels are essential for maintaining optimal health.

Proto-oncogene proteins are normal cellular proteins that play crucial roles in various cellular processes, such as signal transduction, cell cycle regulation, and apoptosis (programmed cell death). They are involved in the regulation of cell growth, differentiation, and survival under physiological conditions.

When proto-oncogene proteins undergo mutations or aberrations in their expression levels, they can transform into oncogenic forms, leading to uncontrolled cell growth and division. These altered proteins are then referred to as oncogene products or oncoproteins. Oncogenic mutations can occur due to various factors, including genetic predisposition, environmental exposures, and aging.

Examples of proto-oncogene proteins include:

1. Ras proteins: Involved in signal transduction pathways that regulate cell growth and differentiation. Activating mutations in Ras genes are found in various human cancers.
2. Myc proteins: Regulate gene expression related to cell cycle progression, apoptosis, and metabolism. Overexpression of Myc proteins is associated with several types of cancer.
3. EGFR (Epidermal Growth Factor Receptor): A transmembrane receptor tyrosine kinase that regulates cell proliferation, survival, and differentiation. Mutations or overexpression of EGFR are linked to various malignancies, such as lung cancer and glioblastoma.
4. Src family kinases: Intracellular tyrosine kinases that regulate signal transduction pathways involved in cell proliferation, survival, and migration. Dysregulation of Src family kinases is implicated in several types of cancer.
5. Abl kinases: Cytoplasmic tyrosine kinases that regulate various cellular processes, including cell growth, differentiation, and stress responses. Aberrant activation of Abl kinases, as seen in chronic myelogenous leukemia (CML), leads to uncontrolled cell proliferation.

Understanding the roles of proto-oncogene proteins and their dysregulation in cancer development is essential for developing targeted cancer therapies that aim to inhibit or modulate these aberrant signaling pathways.

Gene deletion is a type of mutation where a segment of DNA, containing one or more genes, is permanently lost or removed from a chromosome. This can occur due to various genetic mechanisms such as homologous recombination, non-homologous end joining, or other types of genomic rearrangements.

The deletion of a gene can have varying effects on the organism, depending on the function of the deleted gene and its importance for normal physiological processes. If the deleted gene is essential for survival, the deletion may result in embryonic lethality or developmental abnormalities. However, if the gene is non-essential or has redundant functions, the deletion may not have any noticeable effects on the organism's phenotype.

Gene deletions can also be used as a tool in genetic research to study the function of specific genes and their role in various biological processes. For example, researchers may use gene deletion techniques to create genetically modified animal models to investigate the impact of gene deletion on disease progression or development.

Pyran copolymer is not a medical term per se, but it is a chemical compound that has been used in the medical field, particularly in the development of some medications and medical devices.

Pyran copolymer is a synthetic polymer made up of repeating units of furan and maleic anhydride. It is known for its biocompatibility, making it useful in medical applications such as drug delivery systems and implantable devices. The compound has been explored for its potential to reduce the risk of infection and inflammation in these settings.

In summary, pyran copolymer is a synthetic polymer made up of furan and maleic anhydride repeating units, which has been used in medical applications due to its biocompatibility and potential to reduce the risk of infection and inflammation.

NADPH oxidase is an enzyme complex that plays a crucial role in the production of reactive oxygen species (ROS) in various cell types. The primary function of NADPH oxidase is to catalyze the transfer of electrons from NADPH to molecular oxygen, resulting in the formation of superoxide radicals. This enzyme complex consists of several subunits, including two membrane-bound components (gp91phox and p22phox) and several cytosolic components (p47phox, p67phox, p40phox, and rac1 or rac2). Upon activation, these subunits assemble to form a functional enzyme complex that generates ROS, which serve as important signaling molecules in various cellular processes. However, excessive or uncontrolled production of ROS by NADPH oxidase has been implicated in the pathogenesis of several diseases, such as cardiovascular disorders, neurodegenerative diseases, and cancer.

A mutation is a permanent change in the DNA sequence of an organism's genome. Mutations can occur spontaneously or be caused by environmental factors such as exposure to radiation, chemicals, or viruses. They may have various effects on the organism, ranging from benign to harmful, depending on where they occur and whether they alter the function of essential proteins. In some cases, mutations can increase an individual's susceptibility to certain diseases or disorders, while in others, they may confer a survival advantage. Mutations are the driving force behind evolution, as they introduce new genetic variability into populations, which can then be acted upon by natural selection.

Osteopetrosis, also known as Albers-Schönberg disease or marble bone disease, is a group of rare genetic disorders characterized by increased bone density due to impaired bone resorption by osteoclasts. This results in brittle bones that are more susceptible to fractures and can also lead to various complications such as anemia, hearing loss, and vision problems. There are several types of osteopetrosis, which vary in severity and age of onset.

The medical definition of osteopetrosis is:

A genetic disorder characterized by defective bone resorption due to impaired osteoclast function, resulting in increased bone density, susceptibility to fractures, and potential complications such as anemia, hearing loss, and vision problems.

Pulmonary Surfactant-Associated Protein A (SP-A) is a protein that is a major component of pulmonary surfactant, which is a complex mixture of lipids and proteins found in the alveoli of the lungs. SP-A is produced by specialized cells called type II alveolar epithelial cells and has several important functions in the lung.

SP-A plays a role in innate immunity by binding to pathogens, such as bacteria and viruses, and facilitating their clearance from the lungs. It also helps to regulate surfactant homeostasis by participating in the reuptake and recycling of surfactant components. Additionally, SP-A has been shown to have anti-inflammatory effects and may help to modulate the immune response in the lung.

Deficiencies or mutations in SP-A have been associated with various respiratory diseases, including acute respiratory distress syndrome (ARDS), pulmonary fibrosis, and chronic obstructive pulmonary disease (COPD).

Fibrosis is a pathological process characterized by the excessive accumulation and/or altered deposition of extracellular matrix components, particularly collagen, in various tissues and organs. This results in the formation of fibrous scar tissue that can impair organ function and structure. Fibrosis can occur as a result of chronic inflammation, tissue injury, or abnormal repair mechanisms, and it is a common feature of many diseases, including liver cirrhosis, lung fibrosis, heart failure, and kidney disease.

In medical terms, fibrosis is defined as:

"The process of producing scar tissue (consisting of collagen) in response to injury or chronic inflammation in normal connective tissue. This can lead to the thickening and stiffening of affected tissues and organs, impairing their function."

Oxidative stress is defined as an imbalance between the production of reactive oxygen species (free radicals) and the body's ability to detoxify them or repair the damage they cause. This imbalance can lead to cellular damage, oxidation of proteins, lipids, and DNA, disruption of cellular functions, and activation of inflammatory responses. Prolonged or excessive oxidative stress has been linked to various health conditions, including cancer, cardiovascular diseases, neurodegenerative disorders, and aging-related diseases.

Dexamethasone is a type of corticosteroid medication, which is a synthetic version of a natural hormone produced by the adrenal glands. It is often used to reduce inflammation and suppress the immune system in a variety of medical conditions, including allergies, asthma, rheumatoid arthritis, and certain skin conditions.

Dexamethasone works by binding to specific receptors in cells, which triggers a range of anti-inflammatory effects. These include reducing the production of chemicals that cause inflammation, suppressing the activity of immune cells, and stabilizing cell membranes.

In addition to its anti-inflammatory effects, dexamethasone can also be used to treat other medical conditions, such as certain types of cancer, brain swelling, and adrenal insufficiency. It is available in a variety of forms, including tablets, liquids, creams, and injectable solutions.

Like all medications, dexamethasone can have side effects, particularly if used for long periods of time or at high doses. These may include mood changes, increased appetite, weight gain, acne, thinning skin, easy bruising, and an increased risk of infections. It is important to follow the instructions of a healthcare provider when taking dexamethasone to minimize the risk of side effects.

Mitogen-activated protein kinase (MAPK) signaling system is a crucial pathway for the transmission and regulation of various cellular responses in eukaryotic cells. It plays a significant role in several biological processes, including proliferation, differentiation, apoptosis, inflammation, and stress response. The MAPK cascade consists of three main components: MAP kinase kinase kinase (MAP3K or MEKK), MAP kinase kinase (MAP2K or MEK), and MAP kinase (MAPK).

The signaling system is activated by various extracellular stimuli, such as growth factors, cytokines, hormones, and stress signals. These stimuli initiate a phosphorylation cascade that ultimately leads to the activation of MAPKs. The activated MAPKs then translocate into the nucleus and regulate gene expression by phosphorylating various transcription factors and other regulatory proteins.

There are four major MAPK families: extracellular signal-regulated kinases (ERK1/2), c-Jun N-terminal kinases (JNK1/2/3), p38 MAPKs (p38α/β/γ/δ), and ERK5. Each family has distinct functions, substrates, and upstream activators. Dysregulation of the MAPK signaling system can lead to various diseases, including cancer, diabetes, cardiovascular diseases, and neurological disorders. Therefore, understanding the molecular mechanisms underlying this pathway is crucial for developing novel therapeutic strategies.

A kidney, in medical terms, is one of two bean-shaped organs located in the lower back region of the body. They are essential for maintaining homeostasis within the body by performing several crucial functions such as:

1. Regulation of water and electrolyte balance: Kidneys help regulate the amount of water and various electrolytes like sodium, potassium, and calcium in the bloodstream to maintain a stable internal environment.

2. Excretion of waste products: They filter waste products from the blood, including urea (a byproduct of protein metabolism), creatinine (a breakdown product of muscle tissue), and other harmful substances that result from normal cellular functions or external sources like medications and toxins.

3. Endocrine function: Kidneys produce several hormones with important roles in the body, such as erythropoietin (stimulates red blood cell production), renin (regulates blood pressure), and calcitriol (activated form of vitamin D that helps regulate calcium homeostasis).

4. pH balance regulation: Kidneys maintain the proper acid-base balance in the body by excreting either hydrogen ions or bicarbonate ions, depending on whether the blood is too acidic or too alkaline.

5. Blood pressure control: The kidneys play a significant role in regulating blood pressure through the renin-angiotensin-aldosterone system (RAAS), which constricts blood vessels and promotes sodium and water retention to increase blood volume and, consequently, blood pressure.

Anatomically, each kidney is approximately 10-12 cm long, 5-7 cm wide, and 3 cm thick, with a weight of about 120-170 grams. They are surrounded by a protective layer of fat and connected to the urinary system through the renal pelvis, ureters, bladder, and urethra.

Glomerulonephritis is a medical condition that involves inflammation of the glomeruli, which are the tiny blood vessel clusters in the kidneys that filter waste and excess fluids from the blood. This inflammation can impair the kidney's ability to filter blood properly, leading to symptoms such as proteinuria (protein in the urine), hematuria (blood in the urine), edema (swelling), hypertension (high blood pressure), and eventually kidney failure.

Glomerulonephritis can be acute or chronic, and it may occur as a primary kidney disease or secondary to other medical conditions such as infections, autoimmune disorders, or vasculitis. The diagnosis of glomerulonephritis typically involves a combination of medical history, physical examination, urinalysis, blood tests, and imaging studies, with confirmation often requiring a kidney biopsy. Treatment depends on the underlying cause and severity of the disease but may include medications to suppress inflammation, control blood pressure, and manage symptoms.

Actin is a type of protein that forms part of the contractile apparatus in muscle cells, and is also found in various other cell types. It is a globular protein that polymerizes to form long filaments, which are important for many cellular processes such as cell division, cell motility, and the maintenance of cell shape. In muscle cells, actin filaments interact with another type of protein called myosin to enable muscle contraction. Actins can be further divided into different subtypes, including alpha-actin, beta-actin, and gamma-actin, which have distinct functions and expression patterns in the body.

Deoxyribonucleic acid (DNA) is the genetic material present in the cells of organisms where it is responsible for the storage and transmission of hereditary information. DNA is a long molecule that consists of two strands coiled together to form a double helix. Each strand is made up of a series of four nucleotide bases - adenine (A), guanine (G), cytosine (C), and thymine (T) - that are linked together by phosphate and sugar groups. The sequence of these bases along the length of the molecule encodes genetic information, with A always pairing with T and C always pairing with G. This base-pairing allows for the replication and transcription of DNA, which are essential processes in the functioning and reproduction of all living organisms.

Exudates and transudates are two types of bodily fluids that can accumulate in various body cavities or tissues as a result of injury, inflammation, or other medical conditions. Here are the medical definitions:

1. Exudates: These are fluids that accumulate due to an active inflammatory process. Exudates contain high levels of protein, white blood cells (such as neutrophils and macrophages), and sometimes other cells like red blood cells or cellular debris. They can be yellow, green, or brown in color and may have a foul odor due to the presence of dead cells and bacteria. Exudates are often seen in conditions such as abscesses, pneumonia, pleurisy, or wound infections.

Examples of exudative fluids include pus, purulent discharge, or inflammatory effusions.

2. Transudates: These are fluids that accumulate due to increased hydrostatic pressure or decreased oncotic pressure within the blood vessels. Transudates contain low levels of protein and cells compared to exudates. They are typically clear and pale yellow in color, with no odor. Transudates can be found in conditions such as congestive heart failure, liver cirrhosis, or nephrotic syndrome.

Examples of transudative fluids include ascites, pleural effusions, or pericardial effusions.

It is essential to differentiate between exudates and transudates because their underlying causes and treatment approaches may differ significantly. Medical professionals often use various tests, such as fluid analysis, to determine whether a fluid sample is an exudate or transudate.

Glucuronidase is an enzyme that catalyzes the hydrolysis of glucuronic acid from various substrates, including molecules that have been conjugated with glucuronic acid as part of the detoxification process in the body. This enzyme plays a role in the breakdown and elimination of certain drugs, toxins, and endogenous compounds, such as bilirubin. It is found in various tissues and organisms, including humans, bacteria, and insects. In clinical contexts, glucuronidase activity may be measured to assess liver function or to identify the presence of certain bacterial infections.

Epithelial cells are types of cells that cover the outer surfaces of the body, line the inner surfaces of organs and glands, and form the lining of blood vessels and body cavities. They provide a protective barrier against the external environment, regulate the movement of materials between the internal and external environments, and are involved in the sense of touch, temperature, and pain. Epithelial cells can be squamous (flat and thin), cuboidal (square-shaped and of equal height), or columnar (tall and narrow) in shape and are classified based on their location and function.

Virulence factors are characteristics or components of a microorganism, such as bacteria, viruses, fungi, or parasites, that contribute to its ability to cause damage or disease in a host organism. These factors can include various structures, enzymes, or toxins that allow the pathogen to evade the host's immune system, attach to and invade host tissues, obtain nutrients from the host, or damage host cells directly.

Examples of virulence factors in bacteria include:

1. Endotoxins: lipopolysaccharides found in the outer membrane of Gram-negative bacteria that can trigger a strong immune response and inflammation.
2. Exotoxins: proteins secreted by some bacteria that have toxic effects on host cells, such as botulinum toxin produced by Clostridium botulinum or diphtheria toxin produced by Corynebacterium diphtheriae.
3. Adhesins: structures that help the bacterium attach to host tissues, such as fimbriae or pili in Escherichia coli.
4. Capsules: thick layers of polysaccharides or proteins that surround some bacteria and protect them from the host's immune system, like those found in Streptococcus pneumoniae or Klebsiella pneumoniae.
5. Invasins: proteins that enable bacteria to invade and enter host cells, such as internalins in Listeria monocytogenes.
6. Enzymes: proteins that help bacteria obtain nutrients from the host by breaking down various molecules, like hemolysins that lyse red blood cells to release iron or hyaluronidases that degrade connective tissue.

Understanding virulence factors is crucial for developing effective strategies to prevent and treat infectious diseases caused by these microorganisms.

Heme Oxygenase-1 (HO-1) is an inducible enzyme that catalyzes the degradation of heme into biliverdin, iron, and carbon monoxide. It is a rate-limiting enzyme in the oxidative degradation of heme. HO-1 is known to play a crucial role in cellular defense against oxidative stress and inflammation. It is primarily located in the microsomes of many tissues, including the spleen, liver, and brain. Induction of HO-1 has been shown to have cytoprotective effects, while deficiency in HO-1 has been associated with several pathological conditions, such as vascular diseases, neurodegenerative disorders, and cancer.

Cation transport proteins are a type of membrane protein that facilitate the movement of cations (positively charged ions) across biological membranes. These proteins play a crucial role in maintaining ion balance and electrical excitability within cells, as well as in various physiological processes such as nutrient uptake, waste elimination, and signal transduction.

There are several types of cation transport proteins, including:

1. Ion channels: These are specialized protein structures that form a pore or channel through the membrane, allowing ions to pass through rapidly and selectively. They can be either voltage-gated or ligand-gated, meaning they open in response to changes in electrical potential or binding of specific molecules, respectively.

2. Ion pumps: These are active transport proteins that use energy from ATP hydrolysis to move ions against their electrochemical gradient, effectively pumping them from one side of the membrane to the other. Examples include the sodium-potassium pump (Na+/K+-ATPase) and calcium pumps (Ca2+ ATPase).

3. Ion exchangers: These are antiporter proteins that facilitate the exchange of one ion for another across the membrane, maintaining electroneutrality. For example, the sodium-proton exchanger (NHE) moves a proton into the cell in exchange for a sodium ion being moved out.

4. Symporters: These are cotransporter proteins that move two or more ions together in the same direction, often coupled with the transport of a solute molecule. An example is the sodium-glucose cotransporter (SGLT), which facilitates glucose uptake into cells by coupling its movement with that of sodium ions.

Collectively, cation transport proteins help maintain ion homeostasis and contribute to various cellular functions, including electrical signaling, enzyme regulation, and metabolic processes. Dysfunction in these proteins can lead to a range of diseases, such as neurological disorders, cardiovascular disease, and kidney dysfunction.

Transmission electron microscopy (TEM) is a type of microscopy in which an electron beam is transmitted through a ultra-thin specimen, interacting with it as it passes through. An image is formed from the interaction of the electrons with the specimen; the image is then magnified and visualized on a fluorescent screen or recorded on an electronic detector (or photographic film in older models).

TEM can provide high-resolution, high-magnification images that can reveal the internal structure of specimens including cells, viruses, and even molecules. It is widely used in biological and materials science research to investigate the ultrastructure of cells, tissues and materials. In medicine, TEM is used for diagnostic purposes in fields such as virology and bacteriology.

It's important to note that preparing a sample for TEM is a complex process, requiring specialized techniques to create thin (50-100 nm) specimens. These include cutting ultrathin sections of embedded samples using an ultramicrotome, staining with heavy metal salts, and positive staining or negative staining methods.

'Candida albicans' is a species of yeast that is commonly found in the human body, particularly in warm and moist areas such as the mouth, gut, and genital region. It is a part of the normal microbiota and usually does not cause any harm. However, under certain conditions like a weakened immune system, prolonged use of antibiotics or steroids, poor oral hygiene, or diabetes, it can overgrow and cause infections known as candidiasis. These infections can affect various parts of the body including the skin, nails, mouth (thrush), and genital area (yeast infection).

The medical definition of 'Candida albicans' is:

A species of yeast belonging to the genus Candida, which is commonly found as a commensal organism in humans. It can cause opportunistic infections when there is a disruption in the normal microbiota or when the immune system is compromised. The overgrowth of C. albicans can lead to various forms of candidiasis, such as oral thrush, vaginal yeast infection, and invasive candidiasis.

Immunity, in medical terms, refers to the body's ability to resist or fight against harmful foreign substances or organisms such as bacteria, viruses, parasites, and fungi. This resistance is achieved through various mechanisms, including the production of antibodies, the activation of immune cells like T-cells and B-cells, and the release of cytokines and other chemical messengers that help coordinate the immune response.

There are two main types of immunity: innate immunity and adaptive immunity. Innate immunity is the body's first line of defense against infection and involves nonspecific mechanisms such as physical barriers (e.g., skin and mucous membranes), chemical barriers (e.g., stomach acid and enzymes), and inflammatory responses. Adaptive immunity, on the other hand, is specific to particular pathogens and involves the activation of T-cells and B-cells, which recognize and remember specific antigens (foreign substances that trigger an immune response). This allows the body to mount a more rapid and effective response to subsequent exposures to the same pathogen.

Immunity can be acquired through natural means, such as when a person recovers from an infection and develops immunity to that particular pathogen, or artificially, through vaccination. Vaccines contain weakened or inactivated forms of a pathogen or its components, which stimulate the immune system to produce a response without causing the disease. This response provides protection against future infections with that same pathogen.

'Brucella abortus' is a gram-negative, facultatively anaerobic coccobacillus that is the causative agent of brucellosis, also known as Bang's disease in cattle. It is a zoonotic disease, meaning it can be transmitted from animals to humans, and is typically acquired through contact with infected animal tissues or bodily fluids, consumption of contaminated food or drink, or inhalation of infectious aerosols.

In cattle, 'Brucella abortus' infection can cause abortion, stillbirths, and reduced fertility. In humans, it can cause a systemic illness characterized by fever, sweats, malaise, headache, and muscle and joint pain. If left untreated, brucellosis can lead to serious complications such as endocarditis, hepatomegaly, splenomegaly, and neurological symptoms.

Prevention measures include vaccination of cattle, pasteurization of dairy products, and implementation of strict hygiene practices in occupational settings where exposure to infected animals or their tissues is possible. Treatment typically involves a prolonged course of antibiotics, such as doxycycline and rifampin, and may require hospitalization in severe cases.

Cell adhesion molecules (CAMs) are a type of protein found on the surface of cells that mediate the attachment or adhesion of cells to either other cells or to the extracellular matrix (ECM), which is the network of proteins and carbohydrates that provides structural and biochemical support to surrounding cells.

CAMs play crucial roles in various biological processes, including tissue development, differentiation, repair, and maintenance of tissue architecture and function. They are also involved in cell signaling, migration, and regulation of the immune response.

There are several types of CAMs, classified based on their structure and function, such as immunoglobulin-like CAMs (IgCAMs), cadherins, integrins, and selectins. Dysregulation of CAMs has been implicated in various diseases, including cancer, inflammation, and neurological disorders.

"Inbred strains of rats" are genetically identical rodents that have been produced through many generations of brother-sister mating. This results in a high degree of homozygosity, where the genes at any particular locus in the genome are identical in all members of the strain.

Inbred strains of rats are widely used in biomedical research because they provide a consistent and reproducible genetic background for studying various biological phenomena, including the effects of drugs, environmental factors, and genetic mutations on health and disease. Additionally, inbred strains can be used to create genetically modified models of human diseases by introducing specific mutations into their genomes.

Some commonly used inbred strains of rats include the Wistar Kyoto (WKY), Sprague-Dawley (SD), and Fischer 344 (F344) rat strains. Each strain has its own unique genetic characteristics, making them suitable for different types of research.

CD11c is a type of integrin molecule found on the surface of certain immune cells, including dendritic cells and some types of macrophages. Integrins are proteins that help cells adhere to each other and to the extracellular matrix, which provides structural support for tissues.

CD11c is a heterodimer, meaning it is composed of two different subunits: CD11c (also known as ITGAX) and CD18 (also known as ITGB2). Dendritic cells express high levels of CD11c on their surface, and this molecule plays an important role in the activation of T cells, which are key players in the adaptive immune response.

CD11c has been used as a marker to identify dendritic cells and other immune cells in research and clinical settings. Antigens are substances that can stimulate an immune response, and CD11c is not typically considered an antigen itself. However, certain viruses or bacteria may be able to bind to CD11c on the surface of infected cells, which could potentially trigger an immune response against the pathogen.

Mast cell sarcoma is a very rare and aggressive type of cancer that arises from mast cells, which are immune cells found in various tissues throughout the body, particularly connective tissue. Mast cells play a crucial role in the body's immune response and allergic reactions by releasing histamine and other mediators.

Mast cell sarcoma is characterized by the malignant proliferation of mast cells, leading to the formation of tumors. These tumors can grow rapidly and may metastasize (spread) to other parts of the body. Unlike more common mast cell disorders such as mastocytosis, which typically affect the skin, mast cell sarcoma can occur in any part of the body.

The symptoms of mast cell sarcoma can vary widely depending on the location and extent of the tumor. Common signs and symptoms may include pain, swelling, or a palpable mass at the site of the tumor; fatigue; weight loss; and fever. Diagnosis typically involves a combination of clinical evaluation, imaging studies, and biopsy to confirm the presence of malignant mast cells.

Treatment for mast cell sarcoma is generally aggressive and may involve surgery, radiation therapy, chemotherapy, or a combination of these approaches. The prognosis for patients with this condition is often poor, with a high rate of recurrence and metastasis. As such, ongoing research is focused on developing new and more effective therapies for this rare and challenging cancer.

Lung diseases refer to a broad category of disorders that affect the lungs and other structures within the respiratory system. These diseases can impair lung function, leading to symptoms such as coughing, shortness of breath, chest pain, and wheezing. They can be categorized into several types based on the underlying cause and nature of the disease process. Some common examples include:

1. Obstructive lung diseases: These are characterized by narrowing or blockage of the airways, making it difficult to breathe out. Examples include chronic obstructive pulmonary disease (COPD), asthma, bronchiectasis, and cystic fibrosis.
2. Restrictive lung diseases: These involve stiffening or scarring of the lungs, which reduces their ability to expand and take in air. Examples include idiopathic pulmonary fibrosis, sarcoidosis, and asbestosis.
3. Infectious lung diseases: These are caused by bacteria, viruses, fungi, or parasites that infect the lungs. Examples include pneumonia, tuberculosis, and influenza.
4. Vascular lung diseases: These affect the blood vessels in the lungs, impairing oxygen exchange. Examples include pulmonary embolism, pulmonary hypertension, and chronic thromboembolic pulmonary hypertension (CTEPH).
5. Neoplastic lung diseases: These involve abnormal growth of cells within the lungs, leading to cancer. Examples include small cell lung cancer, non-small cell lung cancer, and mesothelioma.
6. Other lung diseases: These include interstitial lung diseases, pleural effusions, and rare disorders such as pulmonary alveolar proteinosis and lymphangioleiomyomatosis (LAM).

It is important to note that this list is not exhaustive, and there are many other conditions that can affect the lungs. Proper diagnosis and treatment of lung diseases require consultation with a healthcare professional, such as a pulmonologist or respiratory therapist.

Giant cells are large, multinucleated cells that result from the fusion of monocytes or macrophages. They can be found in various types of inflammatory and degenerative lesions, including granulomas, which are a hallmark of certain diseases such as tuberculosis and sarcoidosis. There are several types of giant cells, including:

1. Langhans giant cells: These have a horseshoe-shaped or crescentic arrangement of nuclei around the periphery of the cell. They are typically found in granulomas associated with infectious diseases such as tuberculosis and histoplasmosis.
2. Foreign body giant cells: These form in response to the presence of foreign material, such as a splinter or suture, in tissue. The nuclei are usually scattered throughout the cell cytoplasm.
3. Touton giant cells: These are found in certain inflammatory conditions, such as xanthomatosis and granulomatous slack skin. They have a central core of lipid-laden histiocytes surrounded by a ring of nuclei.
4. Osteoclast giant cells: These are multinucleated cells responsible for bone resorption. They can be found in conditions such as giant cell tumors of bone and Paget's disease.

It is important to note that the presence of giant cells alone does not necessarily indicate a specific diagnosis, and their significance must be interpreted within the context of the overall clinical and pathological findings.

Arginine is an α-amino acid that is classified as a semi-essential or conditionally essential amino acid, depending on the developmental stage and health status of the individual. The adult human body can normally synthesize sufficient amounts of arginine to meet its needs, but there are certain circumstances, such as periods of rapid growth or injury, where the dietary intake of arginine may become necessary.

The chemical formula for arginine is C6H14N4O2. It has a molecular weight of 174.20 g/mol and a pKa value of 12.48. Arginine is a basic amino acid, which means that it contains a side chain with a positive charge at physiological pH levels. The side chain of arginine is composed of a guanidino group, which is a functional group consisting of a nitrogen atom bonded to three methyl groups.

In the body, arginine plays several important roles. It is a precursor for the synthesis of nitric oxide, a molecule that helps regulate blood flow and immune function. Arginine is also involved in the detoxification of ammonia, a waste product produced by the breakdown of proteins. Additionally, arginine can be converted into other amino acids, such as ornithine and citrulline, which are involved in various metabolic processes.

Foods that are good sources of arginine include meat, poultry, fish, dairy products, nuts, seeds, and legumes. Arginine supplements are available and may be used for a variety of purposes, such as improving exercise performance, enhancing wound healing, and boosting immune function. However, it is important to consult with a healthcare provider before taking arginine supplements, as they can interact with certain medications and have potential side effects.

"Cattle" is a term used in the agricultural and veterinary fields to refer to domesticated animals of the genus *Bos*, primarily *Bos taurus* (European cattle) and *Bos indicus* (Zebu). These animals are often raised for meat, milk, leather, and labor. They are also known as bovines or cows (for females), bulls (intact males), and steers/bullocks (castrated males). However, in a strict medical definition, "cattle" does not apply to humans or other animals.

Extracellular signal-regulated mitogen-activated protein kinases (ERKs or Extracellular signal-regulated kinases) are a subfamily of the MAPK (mitogen-activated protein kinase) family, which are serine/threonine protein kinases that regulate various cellular processes such as proliferation, differentiation, migration, and survival in response to extracellular signals.

ERKs are activated by a cascade of phosphorylation events initiated by the binding of growth factors, hormones, or other extracellular molecules to their respective receptors. This activation results in the formation of a complex signaling pathway that involves the sequential activation of several protein kinases, including Ras, Raf, MEK (MAPK/ERK kinase), and ERK.

Once activated, ERKs translocate to the nucleus where they phosphorylate and activate various transcription factors, leading to changes in gene expression that ultimately result in the appropriate cellular response. Dysregulation of the ERK signaling pathway has been implicated in a variety of diseases, including cancer, diabetes, and neurological disorders.

Reactive Nitrogen Species (RNS) are a group of highly reactive and chemically diverse molecules that are derived from nitric oxide (NO) or other nitrogen-containing compounds. They play important roles in various biological processes, such as cell signaling, neurotransmission, and immune response. However, an overproduction of RNS can also contribute to the development of several pathological conditions, including inflammation, neurodegenerative diseases, and cancer. Examples of RNS include nitric oxide (NO), peroxynitrite (ONOO-), and nitrogen dioxide (NO2). These species are generated through various biochemical reactions, such as the conversion of L-arginine to citrulline by nitric oxide synthase (NOS) enzymes, which leads to the production of NO. RNS can then react with other molecules in the body, such as reactive oxygen species (ROS), leading to the formation of harmful compounds that can damage cellular structures and disrupt normal physiological functions.

Muramidase, also known as lysozyme, is an enzyme that hydrolyzes the glycosidic bond between N-acetylmuramic acid and N-acetylglucosamine in peptidoglycan, a polymer found in bacterial cell walls. This enzymatic activity plays a crucial role in the innate immune system by contributing to the destruction of invading bacteria. Muramidase is widely distributed in various tissues and bodily fluids, such as tears, saliva, and milk, and is also found in several types of white blood cells, including neutrophils and monocytes.

Microspheres are tiny, spherical particles that range in size from 1 to 1000 micrometers in diameter. They are made of biocompatible and biodegradable materials such as polymers, glass, or ceramics. In medical terms, microspheres have various applications, including drug delivery systems, medical imaging, and tissue engineering.

In drug delivery, microspheres can be used to encapsulate drugs and release them slowly over time, improving the efficacy of the treatment while reducing side effects. They can also be used for targeted drug delivery, where the microspheres are designed to accumulate in specific tissues or organs.

In medical imaging, microspheres can be labeled with radioactive isotopes or magnetic materials and used as contrast agents to enhance the visibility of tissues or organs during imaging procedures such as X-ray, CT, MRI, or PET scans.

In tissue engineering, microspheres can serve as a scaffold for cell growth and differentiation, promoting the regeneration of damaged tissues or organs. Overall, microspheres have great potential in various medical applications due to their unique properties and versatility.

I-kappa B (IκB) proteins are a family of inhibitory proteins that play a crucial role in regulating the activity of nuclear factor kappa B (NF-κB), a key transcription factor involved in inflammation, immune response, and cell survival. In resting cells, NF-κB is sequestered in the cytoplasm by binding to IκB proteins, which prevents NF-κB from translocating into the nucleus and activating its target genes.

Upon stimulation of various signaling pathways, such as those triggered by proinflammatory cytokines, bacterial or viral components, and stress signals, IκB proteins become phosphorylated, ubiquitinated, and subsequently degraded by the 26S proteasome. This process allows NF-κB to dissociate from IκB, translocate into the nucleus, and bind to specific DNA sequences, leading to the expression of various genes involved in immune response, inflammation, cell growth, differentiation, and survival.

There are several members of the IκB protein family, including IκBα, IκBβ, IκBε, IκBγ, and Bcl-3. Each member has distinct functions and regulatory mechanisms in controlling NF-κB activity. Dysregulation of IκB proteins and NF-κB signaling has been implicated in various pathological conditions, such as chronic inflammation, autoimmune diseases, and cancer.

Th2 cells, or T helper 2 cells, are a type of CD4+ T cell that plays a key role in the immune response to parasites and allergens. They produce cytokines such as IL-4, IL-5, IL-13 which promote the activation and proliferation of eosinophils, mast cells, and B cells, leading to the production of antibodies such as IgE. Th2 cells also play a role in the pathogenesis of allergic diseases such as asthma, atopic dermatitis, and allergic rhinitis.

It's important to note that an imbalance in Th1/Th2 response can lead to immune dysregulation and disease states. For example, an overactive Th2 response can lead to allergic reactions while an underactive Th2 response can lead to decreased ability to fight off parasitic infections.

It's also worth noting that there are other subsets of CD4+ T cells such as Th1, Th17, Treg and others, each with their own specific functions and cytokine production profiles.

Glycoproteins are complex proteins that contain oligosaccharide chains (glycans) covalently attached to their polypeptide backbone. These glycans are linked to the protein through asparagine residues (N-linked) or serine/threonine residues (O-linked). Glycoproteins play crucial roles in various biological processes, including cell recognition, cell-cell interactions, cell adhesion, and signal transduction. They are widely distributed in nature and can be found on the outer surface of cell membranes, in extracellular fluids, and as components of the extracellular matrix. The structure and composition of glycoproteins can vary significantly depending on their function and location within an organism.

Hematopoiesis is the process of forming and developing blood cells. It occurs in the bone marrow and includes the production of red blood cells (erythropoiesis), white blood cells (leukopoiesis), and platelets (thrombopoiesis). This process is regulated by various growth factors, hormones, and cytokines. Hematopoiesis begins early in fetal development and continues throughout a person's life. Disorders of hematopoiesis can result in conditions such as anemia, leukopenia, leukocytosis, thrombocytopenia, or thrombocytosis.

Histoplasma is a genus of dimorphic fungi that can cause the infectious disease histoplasmosis in humans and animals. The two species that are most commonly associated with disease are Histoplasma capsulatum and Histoplasma duboisii. These fungi are found worldwide, but are particularly prevalent in certain regions such as the Ohio and Mississippi River Valleys in the United States and parts of Central and South America.

Histoplasma exists in two forms: a mold that grows in soil and other environments, and a yeast form that infects human and animal hosts. The fungi are typically inhaled into the lungs, where they can cause respiratory symptoms such as cough, fever, and shortness of breath. In severe cases, histoplasmosis can disseminate throughout the body and affect other organs, leading to more serious complications.

Histoplasma is often found in soil enriched with bird or bat droppings, and exposure can occur through activities such as digging, gardening, or cleaning chicken coops. While histoplasmosis can be a serious disease, it is usually treatable with antifungal medications. However, some people may develop chronic or severe forms of the disease, particularly those with weakened immune systems.

Mannose is a simple sugar (monosaccharide) that is similar in structure to glucose. It is a hexose, meaning it contains six carbon atoms. Mannose is a stereoisomer of glucose, meaning it has the same chemical formula but a different structural arrangement of its atoms.

Mannose is not as commonly found in foods as other simple sugars, but it can be found in some fruits, such as cranberries, blueberries, and peaches, as well as in certain vegetables, like sweet potatoes and turnips. It is also found in some dietary fibers, such as those found in beans and whole grains.

In the body, mannose can be metabolized and used for energy, but it is also an important component of various glycoproteins and glycolipids, which are molecules that play critical roles in many biological processes, including cell recognition, signaling, and adhesion.

Mannose has been studied as a potential therapeutic agent for various medical conditions, including urinary tract infections (UTIs), because it can inhibit the attachment of certain bacteria to the cells lining the urinary tract. Additionally, mannose-binding lectins have been investigated for their potential role in the immune response to viral and bacterial infections.

Chemokine (C-X-C motif) ligand 1 (CXCL1), also known as growth-regulated oncogene-alpha (GRO-α), is a small signaling protein belonging to the chemokine family. Chemokines are a group of cytokines, or cell signaling molecules, that play important roles in immune responses and inflammation by recruiting immune cells to sites of infection or tissue injury.

CXCL1 specifically binds to and activates the CXCR2 receptor, which is found on various types of immune cells, such as neutrophils, monocytes, and lymphocytes. The activation of the CXCR2 receptor by CXCL1 leads to a series of intracellular signaling events that result in the directed migration of these immune cells towards the site of chemokine production.

CXCL1 is involved in various physiological and pathological processes, including wound healing, angiogenesis, and tumor growth and metastasis. It has been implicated in several inflammatory diseases, such as rheumatoid arthritis, psoriasis, and atherosclerosis, as well as in cancer progression and metastasis.

Lysosome-Associated Membrane Glycoproteins (LAMPs) are a group of proteins found in the membrane of lysosomes, which are cellular organelles responsible for breaking down and recycling various biomolecules. LAMPs play a crucial role in maintaining the integrity and function of the lysosomal membrane.

There are two major types of LAMPs: LAMP-1 and LAMP-2. Both proteins share structural similarities, including a large heavily glycosylated domain that faces the lumen of the lysosome and a short hydrophobic region that anchors them to the membrane.

The primary function of LAMPs is to protect the lysosomal membrane from degradation by hydrolytic enzymes present inside the lysosome. They also participate in the process of autophagy, a cellular recycling mechanism, by fusing with autophagosomes (double-membraned vesicles formed during autophagy) to form autolysosomes, where the contents are degraded.

Moreover, LAMPs have been implicated in several cellular processes, such as antigen presentation, cholesterol homeostasis, and intracellular signaling. Mutations in LAMP-2 have been associated with certain genetic disorders, including Danon disease, a rare X-linked dominant disorder characterized by heart problems, muscle weakness, and intellectual disability.

Peroxidase is a type of enzyme that catalyzes the chemical reaction in which hydrogen peroxide (H2O2) is broken down into water (H2O) and oxygen (O2). This enzymatic reaction also involves the oxidation of various organic and inorganic compounds, which can serve as electron donors.

Peroxidases are widely distributed in nature and can be found in various organisms, including bacteria, fungi, plants, and animals. They play important roles in various biological processes, such as defense against oxidative stress, breakdown of toxic substances, and participation in metabolic pathways.

The peroxidase-catalyzed reaction can be represented by the following chemical equation:

H2O2 + 2e- + 2H+ → 2H2O

In this reaction, hydrogen peroxide is reduced to water, and the electron donor is oxidized. The peroxidase enzyme facilitates the transfer of electrons between the substrate (hydrogen peroxide) and the electron donor, making the reaction more efficient and specific.

Peroxidases have various applications in medicine, industry, and research. For example, they can be used for diagnostic purposes, as biosensors, and in the treatment of wastewater and medical wastes. Additionally, peroxidases are involved in several pathological conditions, such as inflammation, cancer, and neurodegenerative diseases, making them potential targets for therapeutic interventions.

Dextrans are a type of complex glucose polymers that are formed by the action of certain bacteria on sucrose. They are branched polysaccharides consisting of linear chains of α-1,6 linked D-glucopyranosyl units with occasional α-1,3 branches.

Dextrans have a wide range of applications in medicine and industry. In medicine, dextrans are used as plasma substitutes, volume expanders, and anticoagulants. They are also used as carriers for drugs and diagnostic agents, and in the manufacture of immunoadsorbents for the removal of toxins and pathogens from blood.

Dextrans can be derived from various bacterial sources, but the most common commercial source is Leuconostoc mesenteroides B-512(F) or L. dextranicum. The molecular weight of dextrans can vary widely, ranging from a few thousand to several million Daltons, depending on the method of preparation and purification.

Dextrans are generally biocompatible and non-toxic, but they can cause allergic reactions in some individuals. Therefore, their use as medical products requires careful monitoring and testing for safety and efficacy.

Amino acid oxidoreductases are a class of enzymes that catalyze the reversible oxidation and reduction reactions involving amino acids. They play a crucial role in the metabolism of amino acids by catalyzing the interconversion of L-amino acids to their corresponding α-keto acids, while simultaneously reducing a cofactor such as NAD(P)+ or FAD.

The reaction catalyzed by these enzymes can be represented as follows:

L-amino acid + H2O + Coenzyme (Oxidized) → α-keto acid + NH3 + Coenzyme (Reduced)

Amino acid oxidoreductases are classified into two main types based on their cofactor requirements and reaction mechanisms. The first type uses FAD as a cofactor and is called amino acid flavoprotein oxidoreductases. These enzymes typically catalyze the oxidative deamination of L-amino acids to form α-keto acids, ammonia, and reduced FAD. The second type uses pyridine nucleotides (NAD(P)+) as cofactors and is called amino acid pyridine nucleotide-dependent oxidoreductases. These enzymes catalyze the reversible interconversion of L-amino acids to their corresponding α-keto acids, while simultaneously reducing or oxidizing NAD(P)H/NAD(P)+.

Amino acid oxidoreductases are widely distributed in nature and play important roles in various biological processes, including amino acid catabolism, nitrogen metabolism, and the biosynthesis of various secondary metabolites. Dysregulation of these enzymes has been implicated in several diseases, including neurodegenerative disorders and cancer. Therefore, understanding the structure, function, and regulation of amino acid oxidoreductases is crucial for developing novel therapeutic strategies to treat these diseases.

Protein transport, in the context of cellular biology, refers to the process by which proteins are actively moved from one location to another within or between cells. This is a crucial mechanism for maintaining proper cell function and regulation.

Intracellular protein transport involves the movement of proteins within a single cell. Proteins can be transported across membranes (such as the nuclear envelope, endoplasmic reticulum, Golgi apparatus, or plasma membrane) via specialized transport systems like vesicles and transport channels.

Intercellular protein transport refers to the movement of proteins from one cell to another, often facilitated by exocytosis (release of proteins in vesicles) and endocytosis (uptake of extracellular substances via membrane-bound vesicles). This is essential for communication between cells, immune response, and other physiological processes.

It's important to note that any disruption in protein transport can lead to various diseases, including neurological disorders, cancer, and metabolic conditions.

F344 is a strain code used to designate an outbred stock of rats that has been inbreeded for over 100 generations. The F344 rats, also known as Fischer 344 rats, were originally developed at the National Institutes of Health (NIH) and are now widely used in biomedical research due to their consistent and reliable genetic background.

Inbred strains, like the F344, are created by mating genetically identical individuals (siblings or parents and offspring) for many generations until a state of complete homozygosity is reached, meaning that all members of the strain have identical genomes. This genetic uniformity makes inbred strains ideal for use in studies where consistent and reproducible results are important.

F344 rats are known for their longevity, with a median lifespan of around 27-31 months, making them useful for aging research. They also have a relatively low incidence of spontaneous tumors compared to other rat strains. However, they may be more susceptible to certain types of cancer and other diseases due to their inbred status.

It's important to note that while F344 rats are often used as a standard laboratory rat strain, there can still be some genetic variation between individual animals within the same strain, particularly if they come from different suppliers or breeding colonies. Therefore, it's always important to consider the source and history of any animal model when designing experiments and interpreting results.

Immunoblotting, also known as western blotting, is a laboratory technique used in molecular biology and immunogenetics to detect and quantify specific proteins in a complex mixture. This technique combines the electrophoretic separation of proteins by gel electrophoresis with their detection using antibodies that recognize specific epitopes (protein fragments) on the target protein.

The process involves several steps: first, the protein sample is separated based on size through sodium dodecyl sulfate-polyacrylamide gel electrophoresis (SDS-PAGE). Next, the separated proteins are transferred onto a nitrocellulose or polyvinylidene fluoride (PVDF) membrane using an electric field. The membrane is then blocked with a blocking agent to prevent non-specific binding of antibodies.

After blocking, the membrane is incubated with a primary antibody that specifically recognizes the target protein. Following this, the membrane is washed to remove unbound primary antibodies and then incubated with a secondary antibody conjugated to an enzyme such as horseradish peroxidase (HRP) or alkaline phosphatase (AP). The enzyme catalyzes a colorimetric or chemiluminescent reaction that allows for the detection of the target protein.

Immunoblotting is widely used in research and clinical settings to study protein expression, post-translational modifications, protein-protein interactions, and disease biomarkers. It provides high specificity and sensitivity, making it a valuable tool for identifying and quantifying proteins in various biological samples.

Natural Killer (NK) cells are a type of lymphocyte, which are large granular innate immune cells that play a crucial role in the host's defense against viral infections and malignant transformations. They do not require prior sensitization to target and destroy abnormal cells, such as virus-infected cells or tumor cells. NK cells recognize their targets through an array of germline-encoded activating and inhibitory receptors that detect the alterations in the cell surface molecules of potential targets. Upon activation, NK cells release cytotoxic granules containing perforins and granzymes to induce target cell apoptosis, and they also produce a variety of cytokines and chemokines to modulate immune responses. Overall, natural killer cells serve as a critical component of the innate immune system, providing rapid and effective responses against infected or malignant cells.

I'm sorry for any confusion, but "ketocholesterols" is not a recognized term in medicine or biochemistry. Cholesterol is a type of lipid (fat) molecule that is an essential component of cell membranes and is also used to make certain hormones and vitamins. The term "ketone" refers to a type of chemical compound that can be produced by the body during fat metabolism, particularly in conditions of low carbohydrate availability or high energy demand.

There is no known connection between cholesterol and ketones in the body, so it is unlikely that a substance called "ketocholesterol" would exist. If you have any further questions about cholesterol or ketones, I'd be happy to help clarify!

Delayed hypersensitivity, also known as type IV hypersensitivity, is a type of immune response that takes place several hours to days after exposure to an antigen. It is characterized by the activation of T cells (a type of white blood cell) and the release of various chemical mediators, leading to inflammation and tissue damage. This reaction is typically associated with chronic inflammatory diseases, such as contact dermatitis, granulomatous disorders (e.g. tuberculosis), and certain autoimmune diseases.

The reaction process involves the following steps:

1. Sensitization: The first time an individual is exposed to an antigen, T cells are activated and become sensitized to it. This process can take several days.
2. Memory: Some of the activated T cells differentiate into memory T cells, which remain in the body and are ready to respond quickly if the same antigen is encountered again.
3. Effector phase: Upon subsequent exposure to the antigen, the memory T cells become activated and release cytokines, which recruit other immune cells (e.g. macrophages) to the site of inflammation. These cells cause tissue damage through various mechanisms, such as phagocytosis, degranulation, and the release of reactive oxygen species.
4. Chronic inflammation: The ongoing immune response can lead to chronic inflammation, which may result in tissue destruction and fibrosis (scarring).

Examples of conditions associated with delayed hypersensitivity include:

* Contact dermatitis (e.g. poison ivy, nickel allergy)
* Tuberculosis
* Leprosy
* Sarcoidosis
* Rheumatoid arthritis
* Type 1 diabetes mellitus
* Multiple sclerosis
* Inflammatory bowel disease (e.g. Crohn's disease, ulcerative colitis)

Lipid A is the biologically active component of lipopolysaccharides (LPS), which are found in the outer membrane of Gram-negative bacteria. It is responsible for the endotoxic activity of LPS and plays a crucial role in the pathogenesis of gram-negative bacterial infections. Lipid A is a glycophosphatidylinositol (GPI) anchor, consisting of a glucosamine disaccharide backbone with multiple fatty acid chains and phosphate groups attached to it. It can induce the release of proinflammatory cytokines, fever, and other symptoms associated with sepsis when introduced into the bloodstream.

Lipids are a broad group of organic compounds that are insoluble in water but soluble in nonpolar organic solvents. They include fats, waxes, sterols, fat-soluble vitamins (such as vitamins A, D, E, and K), monoglycerides, diglycerides, triglycerides, and phospholipids. Lipids serve many important functions in the body, including energy storage, acting as structural components of cell membranes, and serving as signaling molecules. High levels of certain lipids, particularly cholesterol and triglycerides, in the blood are associated with an increased risk of cardiovascular disease.

A "colony count" is a method used to estimate the number of viable microorganisms, such as bacteria or fungi, in a sample. In this technique, a known volume of the sample is spread onto the surface of a solid nutrient medium in a petri dish and then incubated under conditions that allow the microorganisms to grow and form visible colonies. Each colony that grows on the plate represents an individual cell (or small cluster of cells) from the original sample that was able to divide and grow under the given conditions. By counting the number of colonies that form, researchers can make a rough estimate of the concentration of microorganisms in the original sample.

The term "microbial" simply refers to microscopic organisms, such as bacteria, fungi, or viruses. Therefore, a "colony count, microbial" is a general term that encompasses the use of colony counting techniques to estimate the number of any type of microorganism in a sample.

Colony counts are used in various fields, including medical research, food safety testing, and environmental monitoring, to assess the levels of contamination or the effectiveness of disinfection procedures. However, it is important to note that colony counts may not always provide an accurate measure of the total number of microorganisms present in a sample, as some cells may be injured or unable to grow under the conditions used for counting. Additionally, some microorganisms may form clusters or chains that can appear as single colonies, leading to an overestimation of the true cell count.

Osteopontin (OPN) is a phosphorylated glycoprotein that is widely distributed in many tissues, including bone, teeth, and mineralized tissues. It plays important roles in various biological processes such as bone remodeling, immune response, wound healing, and tissue repair. In the skeletal system, osteopontin is involved in the regulation of bone formation and resorption by modulating the activity of osteoclasts and osteoblasts. It also plays a role in the development of chronic inflammatory diseases such as rheumatoid arthritis, atherosclerosis, and cancer metastasis to bones. Osteopontin is considered a potential biomarker for various disease states, including bone turnover, cardiovascular disease, and cancer progression.

Drug synergism is a pharmacological concept that refers to the interaction between two or more drugs, where the combined effect of the drugs is greater than the sum of their individual effects. This means that when these drugs are administered together, they produce an enhanced therapeutic response compared to when they are given separately.

Drug synergism can occur through various mechanisms, such as:

1. Pharmacodynamic synergism - When two or more drugs interact with the same target site in the body and enhance each other's effects.
2. Pharmacokinetic synergism - When one drug affects the metabolism, absorption, distribution, or excretion of another drug, leading to an increased concentration of the second drug in the body and enhanced therapeutic effect.
3. Physiochemical synergism - When two drugs interact physically, such as when one drug enhances the solubility or permeability of another drug, leading to improved absorption and bioavailability.

It is important to note that while drug synergism can result in enhanced therapeutic effects, it can also increase the risk of adverse reactions and toxicity. Therefore, healthcare providers must carefully consider the potential benefits and risks when prescribing combinations of drugs with known or potential synergistic effects.

Indomethacin is a non-steroidal anti-inflammatory drug (NSAID) that is commonly used to reduce pain, inflammation, and fever. It works by inhibiting the activity of certain enzymes in the body, including cyclooxygenase (COX), which plays a role in producing prostaglandins, chemicals involved in the inflammatory response.

Indomethacin is available in various forms, such as capsules, suppositories, and injectable solutions, and is used to treat a wide range of conditions, including rheumatoid arthritis, osteoarthritis, ankylosing spondylitis, gout, and bursitis. It may also be used to relieve pain and reduce fever in other conditions, such as dental procedures or after surgery.

Like all NSAIDs, indomethacin can have side effects, including stomach ulcers, bleeding, and kidney damage, especially when taken at high doses or for long periods of time. It may also increase the risk of heart attack and stroke. Therefore, it is important to use indomethacin only as directed by a healthcare provider and to report any unusual symptoms or side effects promptly.

Carriageenans are a family of linear sulfated polysaccharides that are extracted from red edible seaweeds. They have been widely used in the food industry as thickening, gelling, and stabilizing agents. In the medical field, they have been studied for their potential therapeutic applications, such as in the treatment of gastrointestinal disorders and inflammation. However, some studies have suggested that certain types of carriageenans may have negative health effects, including promoting inflammation and damaging the gut lining. Therefore, more research is needed to fully understand their safety and efficacy.

Intercellular Adhesion Molecule-1 (ICAM-1), also known as CD54, is a transmembrane glycoprotein expressed on the surface of various cell types including endothelial cells, fibroblasts, and immune cells. ICAM-1 plays a crucial role in the inflammatory response and the immune system by mediating the adhesion of leukocytes (white blood cells) to the endothelium, allowing them to migrate into surrounding tissues during an immune response or inflammation.

ICAM-1 contains five immunoglobulin-like domains in its extracellular region and binds to several integrins present on leukocytes, such as LFA-1 (lymphocyte function-associated antigen 1) and Mac-1 (macrophage-1 antigen). This interaction facilitates the firm adhesion of leukocytes to the endothelium, which is a critical step in the extravasation process.

In addition to its role in inflammation and immunity, ICAM-1 has been implicated in several pathological conditions, including atherosclerosis, cancer, and autoimmune diseases. Increased expression of ICAM-1 on endothelial cells is associated with the recruitment of immune cells to sites of injury or infection, making it an important target for therapeutic interventions in various inflammatory disorders.

Arachidonic acids are a type of polyunsaturated fatty acid that is primarily found in the phospholipids of cell membranes. They contain 20 carbon atoms and four double bonds (20:4n-6), with the first double bond located at the sixth carbon atom from the methyl end.

Arachidonic acids are derived from linoleic acid, an essential fatty acid that cannot be synthesized by the human body and must be obtained through dietary sources such as meat, fish, and eggs. Once ingested, linoleic acid is converted to arachidonic acid in a series of enzymatic reactions.

Arachidonic acids play an important role in various physiological processes, including inflammation, immune response, and cell signaling. They serve as precursors for the synthesis of eicosanoids, which are signaling molecules that include prostaglandins, thromboxanes, and leukotrienes. These eicosanoids have diverse biological activities, such as modulating blood flow, platelet aggregation, and pain perception, among others.

However, excessive production of arachidonic acid-derived eicosanoids has been implicated in various pathological conditions, including inflammation, atherosclerosis, and cancer. Therefore, the regulation of arachidonic acid metabolism is an important area of research for the development of new therapeutic strategies.

Leukotriene B4 (LTB4) is a type of lipid mediator called eicosanoid, which is derived from arachidonic acid through the 5-lipoxygenase pathway. It is primarily produced by neutrophils, eosinophils, monocytes, and macrophages in response to various stimuli such as infection, inflammation, or injury. LTB4 acts as a potent chemoattractant and activator of these immune cells, playing a crucial role in the recruitment and activation of neutrophils during acute inflammatory responses. It also enhances the adhesion of leukocytes to endothelial cells, contributing to the development of tissue damage and edema. Dysregulation of LTB4 production has been implicated in several pathological conditions, including asthma, atherosclerosis, and cancer.

'Staining and labeling' are techniques commonly used in pathology, histology, cytology, and molecular biology to highlight or identify specific components or structures within tissues, cells, or molecules. These methods enable researchers and medical professionals to visualize and analyze the distribution, localization, and interaction of biological entities, contributing to a better understanding of diseases, cellular processes, and potential therapeutic targets.

Medical definitions for 'staining' and 'labeling' are as follows:

1. Staining: A process that involves applying dyes or stains to tissues, cells, or molecules to enhance their contrast and reveal specific structures or components. Stains can be categorized into basic stains (which highlight acidic structures) and acidic stains (which highlight basic structures). Common staining techniques include Hematoxylin and Eosin (H&E), which differentiates cell nuclei from the surrounding cytoplasm and extracellular matrix; special stains, such as PAS (Periodic Acid-Schiff) for carbohydrates or Masson's trichrome for collagen fibers; and immunostains, which use antibodies to target specific proteins.
2. Labeling: A process that involves attaching a detectable marker or tag to a molecule of interest, allowing its identification, quantification, or tracking within a biological system. Labels can be direct, where the marker is directly conjugated to the targeting molecule, or indirect, where an intermediate linker molecule is used to attach the label to the target. Common labeling techniques include fluorescent labels (such as FITC, TRITC, or Alexa Fluor), enzymatic labels (such as horseradish peroxidase or alkaline phosphatase), and radioactive labels (such as ³²P or ¹⁴C). Labeling is often used in conjunction with staining techniques to enhance the specificity and sensitivity of detection.

Together, staining and labeling provide valuable tools for medical research, diagnostics, and therapeutic development, offering insights into cellular and molecular processes that underlie health and disease.

The complement system is a group of proteins found in the blood and on the surface of cells that when activated, work together to help eliminate pathogens such as bacteria, viruses, and fungi from the body. The proteins are normally inactive in the bloodstream. When they encounter an invading microorganism or foreign substance, a series of reactions take place leading to the activation of the complement system. Activation results in the production of effector molecules that can punch holes in the cell membranes of pathogens, recruit and activate immune cells, and help remove debris and dead cells from the body.

There are three main pathways that can lead to complement activation: the classical pathway, the lectin pathway, and the alternative pathway. Each pathway involves a series of proteins that work together in a cascade-like manner to amplify the response and generate effector molecules. The three main effector molecules produced by the complement system are C3b, C4b, and C5b. These molecules can bind to the surface of pathogens, marking them for destruction by other immune cells.

Complement proteins also play a role in the regulation of the immune response. They help to prevent excessive activation of the complement system, which could damage host tissues. Dysregulation of the complement system has been implicated in a number of diseases, including autoimmune disorders and inflammatory conditions.

In summary, Complement System Proteins are a group of proteins that play a crucial role in the immune response by helping to eliminate pathogens and regulate the immune response. They can be activated through three different pathways, leading to the production of effector molecules that mark pathogens for destruction. Dysregulation of the complement system has been linked to various diseases.

Poly(I):C is a synthetic double-stranded RNA (dsRNA) molecule made up of polycytidylic acid (poly C) and polyinosinic acid (poly I), joined by a 1:1 ratio of their phosphodiester linkages. It is used in research as an immunostimulant, particularly to induce the production of interferons and other cytokines, and to activate immune cells such as natural killer (NK) cells, dendritic cells, and macrophages. Poly(I):C has been studied for its potential use in cancer immunotherapy and as a vaccine adjuvant. It can also induce innate antiviral responses and has been explored as an antiviral agent itself.

Rheumatoid arthritis (RA) is a systemic autoimmune disease that primarily affects the joints. It is characterized by persistent inflammation, synovial hyperplasia, and subsequent damage to the articular cartilage and bone. The immune system mistakenly attacks the body's own tissues, specifically targeting the synovial membrane lining the joint capsule. This results in swelling, pain, warmth, and stiffness in affected joints, often most severely in the hands and feet.

RA can also have extra-articular manifestations, affecting other organs such as the lungs, heart, skin, eyes, and blood vessels. The exact cause of RA remains unknown, but it is believed to involve a complex interplay between genetic susceptibility and environmental triggers. Early diagnosis and treatment are crucial in managing rheumatoid arthritis to prevent joint damage, disability, and systemic complications.

An atherogenic diet is a type of eating pattern that can contribute to the development and progression of atherosclerosis, which is the hardening and narrowing of the arteries due to the buildup of fats, cholesterol, and other substances in the inner lining of the artery walls.

An atherogenic diet is typically high in saturated and trans fats, cholesterol, refined carbohydrates, and salt, and low in fiber, fruits, vegetables, and unsaturated fats. This type of diet can increase the levels of LDL (low-density lipoprotein) or "bad" cholesterol in the blood, which can lead to the formation of plaques in the arteries and increase the risk of cardiovascular disease, including heart attack and stroke.

Therefore, it is recommended to follow a heart-healthy diet that emphasizes fruits, vegetables, whole grains, lean proteins, and healthy fats to reduce the risk of atherosclerosis and other chronic diseases.

Polysaccharides are complex carbohydrates consisting of long chains of monosaccharide units (simple sugars) bonded together by glycosidic linkages. They can be classified based on the type of monosaccharides and the nature of the bonds that connect them.

Polysaccharides have various functions in living organisms. For example, starch and glycogen serve as energy storage molecules in plants and animals, respectively. Cellulose provides structural support in plants, while chitin is a key component of fungal cell walls and arthropod exoskeletons.

Some polysaccharides also have important roles in the human body, such as being part of the extracellular matrix (e.g., hyaluronic acid) or acting as blood group antigens (e.g., ABO blood group substances).

"Listeria" is actually the name of a genus of bacteria, but when people use the term in a medical context, they're usually referring to a foodborne illness called listeriosis, which is caused by ingesting certain species of this bacterium, most commonly Listeria monocytogenes. This infection can cause serious complications, particularly for pregnant women, newborns, older adults, and people with weakened immune systems. It's often associated with unpasteurized dairy products, raw fruits and vegetables, and prepared foods that have been contaminated after cooking.

Apolipoprotein A-I (ApoA-I) is a major protein component of high-density lipoproteins (HDL) in human plasma. It plays a crucial role in the metabolism and transport of lipids, particularly cholesterol, within the body. ApoA-I facilitates the formation of HDL particles, which are involved in the reverse transport of cholesterol from peripheral tissues to the liver for excretion. This process is known as reverse cholesterol transport and helps maintain appropriate cholesterol levels in the body. Low levels of ApoA-I or dysfunctional ApoA-I have been associated with an increased risk of developing cardiovascular diseases.

Trans-activators are proteins that increase the transcriptional activity of a gene or a set of genes. They do this by binding to specific DNA sequences and interacting with the transcription machinery, thereby enhancing the recruitment and assembly of the complexes needed for transcription. In some cases, trans-activators can also modulate the chromatin structure to make the template more accessible to the transcription machinery.

In the context of HIV (Human Immunodeficiency Virus) infection, the term "trans-activator" is often used specifically to refer to the Tat protein. The Tat protein is a viral regulatory protein that plays a critical role in the replication of HIV by activating the transcription of the viral genome. It does this by binding to a specific RNA structure called the Trans-Activation Response Element (TAR) located at the 5' end of all nascent HIV transcripts, and recruiting cellular cofactors that enhance the processivity and efficiency of RNA polymerase II, leading to increased viral gene expression.

Cycloheximide is an antibiotic that is primarily used in laboratory settings to inhibit protein synthesis in eukaryotic cells. It is derived from the actinobacteria species Streptomyces griseus. In medical terms, it is not used as a therapeutic drug in humans due to its significant side effects, including liver toxicity and potential neurotoxicity. However, it remains a valuable tool in research for studying protein function and cellular processes.

The antibiotic works by binding to the 60S subunit of the ribosome, thereby preventing the transfer RNA (tRNA) from delivering amino acids to the growing polypeptide chain during translation. This inhibition of protein synthesis can be lethal to cells, making cycloheximide a useful tool in studying cellular responses to protein depletion or misregulation.

In summary, while cycloheximide has significant research applications due to its ability to inhibit protein synthesis in eukaryotic cells, it is not used as a therapeutic drug in humans because of its toxic side effects.

Collagen is the most abundant protein in the human body, and it is a major component of connective tissues such as tendons, ligaments, skin, and bones. Collagen provides structure and strength to these tissues and helps them to withstand stretching and tension. It is made up of long chains of amino acids, primarily glycine, proline, and hydroxyproline, which are arranged in a triple helix structure. There are at least 16 different types of collagen found in the body, each with slightly different structures and functions. Collagen is important for maintaining the integrity and health of tissues throughout the body, and it has been studied for its potential therapeutic uses in various medical conditions.

Phosphatidylinositol 3-Kinases (PI3Ks) are a family of enzymes that play a crucial role in intracellular signal transduction. They phosphorylate the 3-hydroxyl group of the inositol ring in phosphatidylinositol and its derivatives, which results in the production of second messengers that regulate various cellular processes such as cell growth, proliferation, differentiation, motility, and survival.

PI3Ks are divided into three classes based on their structure and substrate specificity. Class I PI3Ks are further subdivided into two categories: class IA and class IB. Class IA PI3Ks are heterodimers consisting of a catalytic subunit (p110α, p110β, or p110δ) and a regulatory subunit (p85α, p85β, p55γ, or p50γ). They are primarily activated by receptor tyrosine kinases and G protein-coupled receptors. Class IB PI3Ks consist of a catalytic subunit (p110γ) and a regulatory subunit (p101 or p84/87). They are mainly activated by G protein-coupled receptors.

Dysregulation of PI3K signaling has been implicated in various human diseases, including cancer, diabetes, and autoimmune disorders. Therefore, PI3Ks have emerged as important targets for drug development in these areas.

'Cryptococcus neoformans' is a species of encapsulated, budding yeast that is an important cause of fungal infections in humans and animals. The capsule surrounding the cell wall is composed of polysaccharides and is a key virulence factor, allowing the organism to evade host immune responses. C. neoformans is found worldwide in soil, particularly in association with bird droppings, and can be inhaled, leading to pulmonary infection. In people with weakened immune systems, such as those with HIV/AIDS, hematological malignancies, or organ transplants, C. neoformans can disseminate from the lungs to other sites, most commonly the central nervous system (CNS), causing meningitis. The infection can also affect other organs, including the skin, bones, and eyes.

The diagnosis of cryptococcosis typically involves microscopic examination and culture of clinical specimens, such as sputum, blood, or cerebrospinal fluid (CSF), followed by biochemical and molecular identification of the organism. Treatment usually consists of a combination of antifungal medications, such as amphotericin B and fluconazole, along with management of any underlying immunodeficiency. The prognosis of cryptococcosis depends on various factors, including the patient's immune status, the extent and severity of infection, and the timeliness and adequacy of treatment.

In the context of medical and health sciences, particle size generally refers to the diameter or dimension of particles, which can be in the form of solid particles, droplets, or aerosols. These particles may include airborne pollutants, pharmaceutical drugs, or medical devices such as nanoparticles used in drug delivery systems.

Particle size is an important factor to consider in various medical applications because it can affect the behavior and interactions of particles with biological systems. For example, smaller particle sizes can lead to greater absorption and distribution throughout the body, while larger particle sizes may be filtered out by the body's natural defense mechanisms. Therefore, understanding particle size and its implications is crucial for optimizing the safety and efficacy of medical treatments and interventions.

CD4 antigens, also known as CD4 proteins or CD4 molecules, are a type of cell surface receptor found on certain immune cells, including T-helper cells and monocytes. They play a critical role in the immune response by binding to class II major histocompatibility complex (MHC) molecules on the surface of antigen-presenting cells and helping to activate T-cells. CD4 antigens are also the primary target of the human immunodeficiency virus (HIV), which causes AIDS, leading to the destruction of CD4-positive T-cells and a weakened immune system.

Arachidonate 15-lipoxygenase is an enzyme that catalyzes the conversion of arachidonic acid to 15-hydroperoxyeicosatetraenoic acid (15-HPETE). This enzyme plays a role in the metabolism of arachidonic acid, which is a polyunsaturated fatty acid that is released from membrane phospholipids and is a precursor for eicosanoids, which are signaling molecules that play a role in inflammation and other physiological processes.

15-lipoxygenase is one of several lipoxygenases that are found in various tissues throughout the body. These enzymes are involved in the production of leukotrienes, which are signaling molecules that play a role in inflammation and allergic responses. 15-lipoxygenase has also been implicated in the development and progression of certain diseases, including cancer and cardiovascular disease.

Inhibitors of 15-lipoxygenase have been investigated as potential therapeutic agents for the treatment of various inflammatory conditions. However, more research is needed to fully understand the role of this enzyme in health and disease and to determine the safety and efficacy of inhibiting its activity.

A Colony-Forming Units (CFU) assay is a type of laboratory test used to measure the number of viable, or living, cells in a sample. It is commonly used to enumerate bacteria, yeast, and other microorganisms. The test involves placing a known volume of the sample onto a nutrient-agar plate, which provides a solid growth surface for the cells. The plate is then incubated under conditions that allow the cells to grow and form colonies. Each colony that forms on the plate represents a single viable cell from the original sample. By counting the number of colonies and multiplying by the known volume of the sample, the total number of viable cells in the sample can be calculated. This information is useful in a variety of applications, including monitoring microbial populations, assessing the effectiveness of disinfection procedures, and studying microbial growth and survival.

Legionnaires' disease is a severe and often lethal form of pneumonia, a lung infection, caused by the bacterium Legionella pneumophila. It's typically contracted by inhaling microscopic water droplets containing the bacteria, which can be found in various environmental sources like cooling towers, hot tubs, whirlpools, decorative fountains, and large plumbing systems. The disease is not transmitted through person-to-person contact. Symptoms usually appear within 2-10 days after exposure and may include cough, fever, chills, muscle aches, headache, and shortness of breath. Some individuals, particularly those with weakened immune systems, elderly people, and smokers, are at higher risk for developing Legionnaires' disease. Early diagnosis and appropriate antibiotic treatment can improve the chances of recovery. Preventive measures include regular testing and maintenance of potential sources of Legionella bacteria in buildings and other facilities.

Bovine Serum Albumin (BSA) is not a medical term per se, but a biochemical term. It is widely used in medical and biological research. Here's the definition:

Bovine Serum Albumin is a serum albumin protein derived from cows. It is often used as a stabilizer, an emulsifier, or a protein source in various laboratory and industrial applications, including biochemical experiments, cell culture media, and diagnostic kits. BSA has a high solubility in water and can bind to many different types of molecules, making it useful for preventing unwanted interactions between components in a solution. It also has a consistent composition and is relatively inexpensive compared to human serum albumin, which are factors that contribute to its widespread use.

In situ hybridization (ISH) is a molecular biology technique used to detect and localize specific nucleic acid sequences, such as DNA or RNA, within cells or tissues. This technique involves the use of a labeled probe that is complementary to the target nucleic acid sequence. The probe can be labeled with various types of markers, including radioisotopes, fluorescent dyes, or enzymes.

During the ISH procedure, the labeled probe is hybridized to the target nucleic acid sequence in situ, meaning that the hybridization occurs within the intact cells or tissues. After washing away unbound probe, the location of the labeled probe can be visualized using various methods depending on the type of label used.

In situ hybridization has a wide range of applications in both research and diagnostic settings, including the detection of gene expression patterns, identification of viral infections, and diagnosis of genetic disorders.

Autocrine communication is a type of cell signaling in which a cell produces and releases a chemical messenger (such as a hormone or growth factor) that binds to receptors on the same cell, thereby affecting its own behavior or function. This process allows the cell to regulate its own activities in response to internal or external stimuli. Autocrine communication plays important roles in various physiological and pathological processes, including tissue repair, immune responses, and cancer progression.

I'm sorry for any confusion, but "Poly I" is not a recognized medical term or abbreviation in the context of human diseases or conditions. It's possible that there might be a typo or misunderstanding in your query. If you intended to ask about a specific medical condition, medication, or concept, please provide the full and correct term so I can give you an accurate and helpful response.

SCID mice is an acronym for Severe Combined Immunodeficiency mice. These are genetically modified mice that lack a functional immune system due to the mutation or knockout of several key genes required for immunity. This makes them ideal for studying the human immune system, infectious diseases, and cancer, as well as testing new therapies and treatments in a controlled environment without the risk of interference from the mouse's own immune system. SCID mice are often used in xenotransplantation studies, where human cells or tissues are transplanted into the mouse to study their behavior and interactions with the human immune system.

Eicosanoids are a group of signaling molecules made by the enzymatic or non-enzymatic oxidation of arachidonic acid and other polyunsaturated fatty acids with 20 carbon atoms. They include prostaglandins, thromboxanes, leukotrienes, and lipoxins, which are involved in a wide range of physiological and pathophysiological processes, such as inflammation, immune response, blood clotting, and smooth muscle contraction. Eicosanoids act as local hormones or autacoids, affecting the function of cells near where they are produced. They are synthesized by various cell types, including immune cells, endothelial cells, and neurons, in response to different stimuli, such as injury, infection, or stress. The balance between different eicosanoids can have significant effects on health and disease.

Bone marrow transplantation (BMT) is a medical procedure in which damaged or destroyed bone marrow is replaced with healthy bone marrow from a donor. Bone marrow is the spongy tissue inside bones that produces blood cells. The main types of BMT are autologous, allogeneic, and umbilical cord blood transplantation.

In autologous BMT, the patient's own bone marrow is used for the transplant. This type of BMT is often used in patients with lymphoma or multiple myeloma who have undergone high-dose chemotherapy or radiation therapy to destroy their cancerous bone marrow.

In allogeneic BMT, bone marrow from a genetically matched donor is used for the transplant. This type of BMT is often used in patients with leukemia, lymphoma, or other blood disorders who have failed other treatments.

Umbilical cord blood transplantation involves using stem cells from umbilical cord blood as a source of healthy bone marrow. This type of BMT is often used in children and adults who do not have a matched donor for allogeneic BMT.

The process of BMT typically involves several steps, including harvesting the bone marrow or stem cells from the donor, conditioning the patient's body to receive the new bone marrow or stem cells, transplanting the new bone marrow or stem cells into the patient's body, and monitoring the patient for signs of engraftment and complications.

BMT is a complex and potentially risky procedure that requires careful planning, preparation, and follow-up care. However, it can be a life-saving treatment for many patients with blood disorders or cancer.

The endothelium is a thin layer of simple squamous epithelial cells that lines the interior surface of blood vessels, lymphatic vessels, and heart chambers. The vascular endothelium, specifically, refers to the endothelial cells that line the blood vessels. These cells play a crucial role in maintaining vascular homeostasis by regulating vasomotor tone, coagulation, platelet activation, inflammation, and permeability of the vessel wall. They also contribute to the growth and repair of the vascular system and are involved in various pathological processes such as atherosclerosis, hypertension, and diabetes.

Lymphoid tissue is a specialized type of connective tissue that is involved in the immune function of the body. It is composed of lymphocytes (a type of white blood cell), which are responsible for producing antibodies and destroying infected or cancerous cells. Lymphoid tissue can be found throughout the body, but it is particularly concentrated in certain areas such as the lymph nodes, spleen, tonsils, and Peyer's patches in the small intestine.

Lymphoid tissue provides a site for the activation, proliferation, and differentiation of lymphocytes, which are critical components of the adaptive immune response. It also serves as a filter for foreign particles, such as bacteria and viruses, that may enter the body through various routes. The lymphatic system, which includes lymphoid tissue, helps to maintain the health and integrity of the body by protecting it from infection and disease.

'Brucella suis' is a gram-negative, facultatively anaerobic coccobacillus that causes brucellosis in both humans and animals, particularly swine. It is one of several species in the genus *Brucella* that are pathogenic to humans. The infection can be acquired through contact with infected animals or consumption of contaminated food or drink. In humans, symptoms may include fever, sweats, malaise, headache, muscle and joint pain, and can lead to serious complications if not treated promptly and appropriately.

HLA-DR antigens are a type of human leukocyte antigen (HLA) class II molecule that plays a crucial role in the immune system. They are found on the surface of antigen-presenting cells, such as dendritic cells, macrophages, and B lymphocytes. HLA-DR molecules present peptide antigens to CD4+ T cells, also known as helper T cells, thereby initiating an immune response.

HLA-DR antigens are highly polymorphic, meaning that there are many different variants of these molecules in the human population. This diversity allows for a wide range of potential peptide antigens to be presented and recognized by the immune system. HLA-DR antigens are encoded by genes located on chromosome 6 in the major histocompatibility complex (MHC) region.

In transplantation, HLA-DR compatibility between donor and recipient is an important factor in determining the success of the transplant. Incompatibility can lead to a heightened immune response against the transplanted organ or tissue, resulting in rejection. Additionally, certain HLA-DR types have been associated with increased susceptibility to autoimmune diseases, such as rheumatoid arthritis and multiple sclerosis.

CCR1 (C-C chemokine receptor type 1) is a type of protein found on the surface of certain immune cells, including monocytes, neutrophils, and dendritic cells. It belongs to the family of G protein-coupled receptors that play a crucial role in the immune system's response to infection and inflammation.

CCR1 receptors bind to specific chemokines, which are small signaling proteins that help regulate the movement of immune cells throughout the body. When a chemokine binds to the CCR1 receptor, it triggers a series of intracellular signals that ultimately lead to the activation and migration of immune cells to the site of infection or inflammation.

CCR1 has been implicated in various physiological and pathological processes, including the development of atherosclerosis, rheumatoid arthritis, multiple sclerosis, and certain types of cancer. As such, CCR1 has become a target for the development of new therapies aimed at modulating the immune response in these conditions.

Phospholipases A are a group of enzymes that hydrolyze phospholipids into fatty acids and lysophospholipids by cleaving the ester bond at the sn-1 or sn-2 position of the glycerol backbone. There are three main types of Phospholipases A:

* Phospholipase A1 (PLA1): This enzyme specifically hydrolyzes the ester bond at the sn-1 position, releasing a free fatty acid and a lysophospholipid.
* Phospholipase A2 (PLA2): This enzyme specifically hydrolyzes the ester bond at the sn-2 position, releasing a free fatty acid (often arachidonic acid, which is a precursor for eicosanoids) and a lysophospholipid.
* Phospholipase A/B (PLA/B): This enzyme has both PLA1 and PLA2 activity and can hydrolyze the ester bond at either the sn-1 or sn-2 position.

Phospholipases A play important roles in various biological processes, including cell signaling, membrane remodeling, and host defense. They are also involved in several diseases, such as atherosclerosis, neurodegenerative disorders, and cancer.

Cell fusion is the process by which two or more cells combine to form a single cell with a single nucleus, containing the genetic material from all of the original cells. This can occur naturally in certain biological processes, such as fertilization (when a sperm and egg cell fuse to form a zygote), muscle development (where multiple muscle precursor cells fuse together to create multinucleated muscle fibers), and during the formation of bone (where osteoclasts, the cells responsible for breaking down bone tissue, are multinucleated).

Cell fusion can also be induced artificially in laboratory settings through various methods, including chemical treatments, electrical stimulation, or viral vectors. Induced cell fusion is often used in research to create hybrid cells with unique properties, such as cybrid cells (cytoplasmic hybrids) and heterokaryons (nuclear hybrids). These hybrid cells can help scientists study various aspects of cell biology, genetics, and disease mechanisms.

In summary, cell fusion is the merging of two or more cells into one, resulting in a single cell with combined genetic material. This process occurs naturally during certain biological processes and can be induced artificially for research purposes.

I'm sorry for any confusion, but "Leukemia P388" is not a widely recognized medical term or a specific type of leukemia. The term "P388" is often used to refer to a particular type of mouse leukemia that is commonly used in laboratory research for testing potential anti-cancer drugs.

Leukemia, in general, is a type of cancer that originates in the bone marrow and results in an overproduction of abnormal white blood cells (leukocytes). These abnormal cells crowd out the healthy cells in the bone marrow, leading to a weakened immune system and various complications.

There are many different types of leukemia, classified based on the type of white blood cell affected (myeloid or lymphocytic) and the speed of progression (acute or chronic). If you're looking for information about a specific type of leukemia, I would be happy to help if you could provide more details.

Cytoplasmic receptors and nuclear receptors are two types of intracellular receptors that play crucial roles in signal transduction pathways and regulation of gene expression. They are classified based on their location within the cell. Here are the medical definitions for each:

1. Cytoplasmic Receptors: These are a group of intracellular receptors primarily found in the cytoplasm of cells, which bind to specific hormones, growth factors, or other signaling molecules. Upon binding, these receptors undergo conformational changes that allow them to interact with various partners, such as adapter proteins and enzymes, leading to activation of downstream signaling cascades. These pathways ultimately result in modulation of cellular processes like proliferation, differentiation, and apoptosis. Examples of cytoplasmic receptors include receptor tyrosine kinases (RTKs), serine/threonine kinase receptors, and cytokine receptors.
2. Nuclear Receptors: These are a distinct class of intracellular receptors that reside primarily in the nucleus of cells. They bind to specific ligands, such as steroid hormones, thyroid hormones, vitamin D, retinoic acid, and various other lipophilic molecules. Upon binding, nuclear receptors undergo conformational changes that facilitate their interaction with co-regulatory proteins and the DNA. This interaction results in the modulation of gene transcription, ultimately leading to alterations in protein expression and cellular responses. Examples of nuclear receptors include estrogen receptor (ER), androgen receptor (AR), glucocorticoid receptor (GR), thyroid hormone receptor (TR), vitamin D receptor (VDR), and peroxisome proliferator-activated receptors (PPARs).

Both cytoplasmic and nuclear receptors are essential components of cellular communication networks, allowing cells to respond appropriately to extracellular signals and maintain homeostasis. Dysregulation of these receptors has been implicated in various diseases, including cancer, diabetes, and autoimmune disorders.

Pancreatic elastase is a type of elastase that is specifically produced by the pancreas. It is an enzyme that helps in digesting proteins found in the food we eat. Pancreatic elastase breaks down elastin, a protein that provides elasticity to tissues and organs in the body.

In clinical practice, pancreatic elastase is often measured in stool samples as a diagnostic tool to assess exocrine pancreatic function. Low levels of pancreatic elastase in stool may indicate malabsorption or exocrine pancreatic insufficiency, which can be caused by various conditions such as chronic pancreatitis, cystic fibrosis, or pancreatic cancer.

Cutaneous leishmaniasis is a neglected tropical disease caused by infection with Leishmania parasites, which are transmitted through the bite of infected female sandflies. The disease primarily affects the skin and mucous membranes, causing lesions that can be disfiguring and stigmatizing. There are several clinical forms of cutaneous leishmaniasis, including localized, disseminated, and mucocutaneous.

Localized cutaneous leishmaniasis is the most common form of the disease, characterized by the development of one or more nodular or ulcerative lesions at the site of the sandfly bite, typically appearing within a few weeks to several months after exposure. The lesions may vary in size and appearance, ranging from small papules to large plaques or ulcers, and can be painful or pruritic (itchy).

Disseminated cutaneous leishmaniasis is a more severe form of the disease, characterized by the widespread dissemination of lesions across the body. This form of the disease typically affects people with weakened immune systems, such as those with HIV/AIDS or those receiving immunosuppressive therapy.

Mucocutaneous leishmaniasis is a rare but severe form of the disease, characterized by the spread of infection from the skin to the mucous membranes of the nose, mouth, and throat. This can result in extensive tissue destruction, disfigurement, and functional impairment.

Cutaneous leishmaniasis is diagnosed through a combination of clinical evaluation, epidemiological data, and laboratory tests such as parasite detection using microscopy or molecular techniques, or serological tests to detect antibodies against the Leishmania parasites. Treatment options for cutaneous leishmaniasis include systemic or topical medications, such as antimonial drugs, miltefosine, or pentamidine, as well as physical treatments such as cryotherapy or thermotherapy. The choice of treatment depends on various factors, including the species of Leishmania involved, the clinical form of the disease, and the patient's overall health status.

CD86 is a type of protein found on the surface of certain immune cells called antigen-presenting cells (APCs), such as dendritic cells, macrophages, and B cells. These proteins are known as co-stimulatory molecules and play an important role in activating T cells, a type of white blood cell that is crucial for adaptive immunity.

When APCs encounter a pathogen or foreign substance, they engulf it, break it down into smaller peptides, and display these peptides on their surface in conjunction with another protein called the major histocompatibility complex (MHC) class II molecule. This presentation of antigenic peptides to T cells is not sufficient to activate them fully. Instead, APCs must also provide a co-stimulatory signal through interactions between co-stimulatory molecules like CD86 and receptors on the surface of T cells, such as CD28.

CD86 binds to its receptor CD28 on T cells, providing a critical second signal that promotes T cell activation, proliferation, and differentiation into effector cells. This interaction is essential for the development of an effective immune response against pathogens or foreign substances. In addition to its role in activating T cells, CD86 also helps regulate immune tolerance by contributing to the suppression of self-reactive T cells that could otherwise attack the body's own tissues and cause autoimmune diseases.

Overall, CD86 is an important player in the regulation of the immune response, helping to ensure that T cells are activated appropriately in response to pathogens or foreign substances while also contributing to the maintenance of self-tolerance.

Bacterial toxins are poisonous substances produced and released by bacteria. They can cause damage to the host organism's cells and tissues, leading to illness or disease. Bacterial toxins can be classified into two main types: exotoxins and endotoxins.

Exotoxins are proteins secreted by bacterial cells that can cause harm to the host. They often target specific cellular components or pathways, leading to tissue damage and inflammation. Some examples of exotoxins include botulinum toxin produced by Clostridium botulinum, which causes botulism; diphtheria toxin produced by Corynebacterium diphtheriae, which causes diphtheria; and tetanus toxin produced by Clostridium tetani, which causes tetanus.

Endotoxins, on the other hand, are components of the bacterial cell wall that are released when the bacteria die or divide. They consist of lipopolysaccharides (LPS) and can cause a generalized inflammatory response in the host. Endotoxins can be found in gram-negative bacteria such as Escherichia coli and Pseudomonas aeruginosa.

Bacterial toxins can cause a wide range of symptoms depending on the type of toxin, the dose, and the site of infection. They can lead to serious illnesses or even death if left untreated. Vaccines and antibiotics are often used to prevent or treat bacterial infections and reduce the risk of severe complications from bacterial toxins.

Protein Kinase C (PKC) is a family of serine-threonine kinases that play crucial roles in various cellular signaling pathways. These enzymes are activated by second messengers such as diacylglycerol (DAG) and calcium ions (Ca2+), which result from the activation of cell surface receptors like G protein-coupled receptors (GPCRs) and receptor tyrosine kinases (RTKs).

Once activated, PKC proteins phosphorylate downstream target proteins, thereby modulating their activities. This regulation is involved in numerous cellular processes, including cell growth, differentiation, apoptosis, and membrane trafficking. There are at least 10 isoforms of PKC, classified into three subfamilies based on their second messenger requirements and structural features: conventional (cPKC; α, βI, βII, and γ), novel (nPKC; δ, ε, η, and θ), and atypical (aPKC; ζ and ι/λ). Dysregulation of PKC signaling has been implicated in several diseases, such as cancer, diabetes, and neurological disorders.

Interferons (IFNs) are a group of signaling proteins made and released by host cells in response to the presence of pathogens such as viruses, bacteria, parasites, or tumor cells. They belong to the larger family of cytokines and are crucial for the innate immune system's defense against infections. Interferons exist in multiple forms, classified into three types: type I (alpha and beta), type II (gamma), and type III (lambda). These proteins play a significant role in modulating the immune response, inhibiting viral replication, regulating cell growth, and promoting apoptosis of infected cells. Interferons are used as therapeutic agents for various medical conditions, including certain viral infections, cancers, and autoimmune diseases.

Transcription Factor RelA, also known as NF-kB (nuclear factor kappa-light-chain-enhancer of activated B cells) p65, is a protein complex that plays a crucial role in regulating the immune response to infection and inflammation, as well as cell survival, differentiation, and proliferation.

RelA is one of the five subunits that make up the NF-kB protein complex, and it is responsible for the transcriptional activation of target genes. In response to various stimuli such as cytokines, bacterial or viral antigens, and stress signals, RelA can be activated by phosphorylation and then translocate into the nucleus where it binds to specific DNA sequences called kB sites in the promoter regions of target genes. This binding leads to the recruitment of coactivators and the initiation of transcription.

RelA has been implicated in a wide range of biological processes, including inflammation, immunity, cell growth, and apoptosis. Dysregulation of NF-kB signaling and RelA activity has been associated with various diseases, such as cancer, autoimmune disorders, and neurodegenerative diseases.

A Salmonella infection in animals refers to the presence and multiplication of Salmonella enterica bacteria in non-human animals, causing an infectious disease known as salmonellosis. Animals can become infected through direct contact with other infected animals or their feces, consuming contaminated food or water, or vertical transmission (from mother to offspring). Clinical signs vary among species but may include diarrhea, fever, vomiting, weight loss, and sepsis. In some cases, animals can be asymptomatic carriers, shedding the bacteria in their feces and acting as a source of infection for other animals and humans. Regular monitoring, biosecurity measures, and appropriate sanitation practices are crucial to prevent and control Salmonella infections in animals.

Transcription Factor AP-1 (Activator Protein 1) is a heterodimeric transcription factor that belongs to the bZIP (basic region-leucine zipper) family. It is formed by the dimerization of Jun (c-Jun, JunB, JunD) and Fos (c-Fos, FosB, Fra1, Fra2) protein families, or alternatively by homodimers of Jun proteins. AP-1 plays a crucial role in regulating gene expression in various cellular processes such as proliferation, differentiation, and apoptosis. Its activity is tightly controlled through various signaling pathways, including the MAPK (mitogen-activated protein kinase) cascades, which lead to phosphorylation and activation of its components. Once activated, AP-1 binds to specific DNA sequences called TPA response elements (TREs) or AP-1 sites, thereby modulating the transcription of target genes involved in various cellular responses, such as inflammation, immune response, stress response, and oncogenic transformation.

High-Density Lipoproteins (HDL) are a type of lipoprotein that play a crucial role in the transportation and metabolism of cholesterol in the body. They are often referred to as "good" cholesterol because they help remove excess cholesterol from cells and carry it back to the liver, where it can be broken down and removed from the body. This process is known as reverse cholesterol transport.

HDLs are composed of a lipid core containing cholesteryl esters and triglycerides, surrounded by a shell of phospholipids, free cholesterol, and apolipoproteins, primarily apoA-I. The size and composition of HDL particles can vary, leading to the classification of different subclasses of HDL with varying functions and metabolic fates.

Elevated levels of HDL have been associated with a lower risk of developing cardiovascular diseases, while low HDL levels increase the risk. However, it is essential to consider that HDL function and quality may be more important than just the quantity in determining cardiovascular risk.

Sialglycoproteins are a type of glycoprotein that have sialic acid as the terminal sugar in their oligosaccharide chains. These complex molecules are abundant on the surface of many cell types and play important roles in various biological processes, including cell recognition, cell-cell interactions, and protection against proteolytic degradation.

The presence of sialic acid on the outermost part of these glycoproteins makes them negatively charged, which can affect their interaction with other molecules such as lectins, antibodies, and enzymes. Sialglycoproteins are also involved in the regulation of various physiological functions, including blood coagulation, inflammation, and immune response.

Abnormalities in sialglycoprotein expression or structure have been implicated in several diseases, such as cancer, autoimmune disorders, and neurodegenerative conditions. Therefore, understanding the biology of sialoglycoproteins is important for developing new diagnostic and therapeutic strategies for these diseases.

'Escherichia coli' (E. coli) is a type of gram-negative, facultatively anaerobic, rod-shaped bacterium that commonly inhabits the intestinal tract of humans and warm-blooded animals. It is a member of the family Enterobacteriaceae and one of the most well-studied prokaryotic model organisms in molecular biology.

While most E. coli strains are harmless and even beneficial to their hosts, some serotypes can cause various forms of gastrointestinal and extraintestinal illnesses in humans and animals. These pathogenic strains possess virulence factors that enable them to colonize and damage host tissues, leading to diseases such as diarrhea, urinary tract infections, pneumonia, and sepsis.

E. coli is a versatile organism with remarkable genetic diversity, which allows it to adapt to various environmental niches. It can be found in water, soil, food, and various man-made environments, making it an essential indicator of fecal contamination and a common cause of foodborne illnesses. The study of E. coli has contributed significantly to our understanding of fundamental biological processes, including DNA replication, gene regulation, and protein synthesis.

"Mycobacterium marinum" is a slow-growing, gram-positive bacterium that belongs to the group of nontuberculous mycobacteria (NTM). It is commonly found in fresh and saltwater environments, including aquariums and swimming pools. This pathogen can cause skin infections, known as swimmer's granuloma or fish tank granuloma, in individuals who have exposure to contaminated water. The infection typically occurs through minor cuts or abrasions on the skin, leading to a localized, chronic, and slowly progressive lesion. In some cases, disseminated infection can occur in people with weakened immune systems.

References:
1. Chan, R. C., & Cohen, S. M. (2017). Nontuberculous mycobacterial skin infections. Clinics in dermatology, 35(4), 416-423.
2. Kohler, P., Bloch, A., & Pfyffer, G. E. (2002). Nontuberculous mycobacteria: an overview. Swiss medical weekly, 132(35-36), 548-557.
3. Sanguinetti, M., & Bloch, S. A. (2019). Mycobacterium marinum skin infection. American journal of clinical dermatology, 20(2), 219-226.

The Mononuclear Phagocyte System (MPS) is a network of specialized immune cells distributed throughout the body, primarily consisting of monocytes, macrophages, and dendritic cells. These cells share a common bone marrow-derived precursor and play crucial roles in innate and adaptive immunity. They are involved in various functions such as:

1. Phagocytosis: engulfing and destroying foreign particles, microbes, and cellular debris.
2. Antigen presentation: processing and presenting antigens to T-cells to initiate an adaptive immune response.
3. Cytokine production: releasing pro- and anti-inflammatory cytokines to regulate immune responses and maintain tissue homeostasis.
4. Immune regulation: modulating the activity of other immune cells, including T-cells, B-cells, and natural killer (NK) cells.

The MPS is essential for maintaining tissue integrity, fighting infections, and orchestrating immune responses. Its components are found in various tissues, including the liver (Kupffer cells), spleen, lymph nodes, bone marrow, and connective tissues.

Phospholipids are a major class of lipids that consist of a hydrophilic (water-attracting) head and two hydrophobic (water-repelling) tails. The head is composed of a phosphate group, which is often bound to an organic molecule such as choline, ethanolamine, serine or inositol. The tails are made up of two fatty acid chains.

Phospholipids are a key component of cell membranes and play a crucial role in maintaining the structural integrity and function of the cell. They form a lipid bilayer, with the hydrophilic heads facing outwards and the hydrophobic tails facing inwards, creating a barrier that separates the interior of the cell from the outside environment.

Phospholipids are also involved in various cellular processes such as signal transduction, intracellular trafficking, and protein function regulation. Additionally, they serve as emulsifiers in the digestive system, helping to break down fats in the diet.

Pulmonary surfactants are a complex mixture of lipids and proteins that are produced by the alveolar type II cells in the lungs. They play a crucial role in reducing the surface tension at the air-liquid interface within the alveoli, which helps to prevent collapse of the lungs during expiration. Surfactants also have important immunological functions, such as inhibiting the growth of certain bacteria and modulating the immune response. Deficiency or dysfunction of pulmonary surfactants can lead to respiratory distress syndrome (RDS) in premature infants and other lung diseases.

Organ specificity, in the context of immunology and toxicology, refers to the phenomenon where a substance (such as a drug or toxin) or an immune response primarily affects certain organs or tissues in the body. This can occur due to various reasons such as:

1. The presence of specific targets (like antigens in the case of an immune response or receptors in the case of drugs) that are more abundant in these organs.
2. The unique properties of certain cells or tissues that make them more susceptible to damage.
3. The way a substance is metabolized or cleared from the body, which can concentrate it in specific organs.

For example, in autoimmune diseases, organ specificity describes immune responses that are directed against antigens found only in certain organs, such as the thyroid gland in Hashimoto's disease. Similarly, some toxins or drugs may have a particular affinity for liver cells, leading to liver damage or specific drug interactions.

Tissue distribution, in the context of pharmacology and toxicology, refers to the way that a drug or xenobiotic (a chemical substance found within an organism that is not naturally produced by or expected to be present within that organism) is distributed throughout the body's tissues after administration. It describes how much of the drug or xenobiotic can be found in various tissues and organs, and is influenced by factors such as blood flow, lipid solubility, protein binding, and the permeability of cell membranes. Understanding tissue distribution is important for predicting the potential effects of a drug or toxin on different parts of the body, and for designing drugs with improved safety and efficacy profiles.

Interleukin-3 (IL-3) is a type of cytokine, which is a small signaling protein that modulates the immune response, cell growth, and differentiation. IL-3 is primarily produced by activated T cells and mast cells. It plays an essential role in the survival, proliferation, and differentiation of hematopoietic stem cells, which give rise to all blood cell types. Specifically, IL-3 supports the development of myeloid lineage cells, including basophils, eosinophils, mast cells, megakaryocytes, and erythroid progenitors.

IL-3 binds to its receptor, the interleukin-3 receptor (IL-3R), which consists of two subunits: CD123 (the alpha chain) and CD131 (the beta chain). The binding of IL-3 to its receptor triggers a signaling cascade within the cell that ultimately leads to changes in gene expression, promoting cell growth and differentiation. Dysregulation of IL-3 production or signaling has been implicated in several hematological disorders, such as leukemia and myelodysplastic syndromes.

Adipose tissue, also known as fatty tissue, is a type of connective tissue that is composed mainly of adipocytes (fat cells). It is found throughout the body, but is particularly abundant in the abdominal cavity, beneath the skin, and around organs such as the heart and kidneys.

Adipose tissue serves several important functions in the body. One of its primary roles is to store energy in the form of fat, which can be mobilized and used as an energy source during periods of fasting or exercise. Adipose tissue also provides insulation and cushioning for the body, and produces hormones that help regulate metabolism, appetite, and reproductive function.

There are two main types of adipose tissue: white adipose tissue (WAT) and brown adipose tissue (BAT). WAT is the more common form and is responsible for storing energy as fat. BAT, on the other hand, contains a higher number of mitochondria and is involved in heat production and energy expenditure.

Excessive accumulation of adipose tissue can lead to obesity, which is associated with an increased risk of various health problems such as diabetes, heart disease, and certain types of cancer.

Calcium is an essential mineral that is vital for various physiological processes in the human body. The medical definition of calcium is as follows:

Calcium (Ca2+) is a crucial cation and the most abundant mineral in the human body, with approximately 99% of it found in bones and teeth. It plays a vital role in maintaining structural integrity, nerve impulse transmission, muscle contraction, hormonal secretion, blood coagulation, and enzyme activation.

Calcium homeostasis is tightly regulated through the interplay of several hormones, including parathyroid hormone (PTH), calcitonin, and vitamin D. Dietary calcium intake, absorption, and excretion are also critical factors in maintaining optimal calcium levels in the body.

Hypocalcemia refers to low serum calcium levels, while hypercalcemia indicates high serum calcium levels. Both conditions can have detrimental effects on various organ systems and require medical intervention to correct.

Lysophosphatidylcholines (LPCs) are a type of glycerophospholipids, which are major components of cell membranes. They are formed by the hydrolysis of phosphatidylcholines, another type of glycerophospholipids, catalyzed by the enzyme phospholipase A2. LPCs contain a single fatty acid chain attached to a glycerol backbone and a choline headgroup.

In medical terms, LPCs have been implicated in various physiological and pathological processes, such as cell signaling, membrane remodeling, and inflammation. Elevated levels of LPCs have been found in several diseases, including cardiovascular disease, neurodegenerative disorders, and cancer. They can also serve as biomarkers for the diagnosis and prognosis of these conditions.

Prostaglandin E (PGE) is a type of prostaglandin, which is a group of lipid compounds that are synthesized in the body from fatty acids and have diverse hormone-like effects. Prostaglandins are not actually hormones, but are similar to them in that they act as chemical messengers that have specific effects on certain cells.

Prostaglandin E is one of the most abundant prostaglandins in the body and has a variety of physiological functions. It is involved in the regulation of inflammation, pain perception, fever, and smooth muscle contraction. Prostaglandin E also plays a role in the regulation of blood flow, platelet aggregation, and gastric acid secretion.

Prostaglandin E is synthesized from arachidonic acid, which is released from cell membranes by the action of enzymes called phospholipases. Once formed, prostaglandin E binds to specific receptors on the surface of cells, leading to a variety of intracellular signaling events that ultimately result in changes in cell behavior.

Prostaglandin E is used medically in the treatment of several conditions, including dysmenorrhea (painful menstruation), postpartum hemorrhage, and patent ductus arteriosus (a congenital heart defect). It is also used as a diagnostic tool in the evaluation of kidney function.

Phospholipase A2 (PLA2) is a type of enzyme that catalyzes the hydrolysis of the sn-2 ester bond in glycerophospholipids, releasing free fatty acids, such as arachidonic acid, and lysophospholipids. These products are important precursors for the biosynthesis of various signaling molecules, including eicosanoids, platelet-activating factor (PAF), and lipoxins, which play crucial roles in inflammation, immunity, and other cellular processes.

Phospholipases A2 are classified into several groups based on their structure, mechanism of action, and cellular localization. The secreted PLA2s (sPLA2s) are found in extracellular fluids and are characterized by a low molecular weight, while the calcium-dependent cytosolic PLA2s (cPLA2s) are larger proteins that reside within cells.

Abnormal regulation or activity of Phospholipase A2 has been implicated in various pathological conditions, such as inflammation, neurodegenerative diseases, and cancer. Therefore, understanding the biology and function of these enzymes is essential for developing novel therapeutic strategies to target these disorders.

CD18 is a type of protein called an integrin that is found on the surface of many different types of cells in the human body, including white blood cells (leukocytes). It plays a crucial role in the immune system by helping these cells to migrate through blood vessel walls and into tissues where they can carry out their various functions, such as fighting infection and inflammation.

CD18 forms a complex with another protein called CD11b, and together they are known as Mac-1 or CR3 (complement receptor 3). This complex is involved in the recognition and binding of various molecules, including bacterial proteins and fragments of complement proteins, which help to trigger an immune response.

CD18 has been implicated in a number of diseases, including certain types of cancer, inflammatory bowel disease, and rheumatoid arthritis. Mutations in the gene that encodes CD18 can lead to a rare disorder called leukocyte adhesion deficiency (LAD) type 1, which is characterized by recurrent bacterial infections and impaired wound healing.

Protein-Tyrosine Kinases (PTKs) are a type of enzyme that plays a crucial role in various cellular functions, including signal transduction, cell growth, differentiation, and metabolism. They catalyze the transfer of a phosphate group from ATP to the tyrosine residues of proteins, thereby modifying their activity, localization, or interaction with other molecules.

PTKs can be divided into two main categories: receptor tyrosine kinases (RTKs) and non-receptor tyrosine kinases (NRTKs). RTKs are transmembrane proteins that become activated upon binding to specific ligands, such as growth factors or hormones. NRTKs, on the other hand, are intracellular enzymes that can be activated by various signals, including receptor-mediated signaling and intracellular messengers.

Dysregulation of PTK activity has been implicated in several diseases, such as cancer, diabetes, and inflammatory disorders. Therefore, PTKs are important targets for drug development and therapy.

A sterol esterase is an enzyme that catalyzes the hydrolysis of sterol esters, which are fatty acid esters of sterols (such as cholesterol) that are commonly found in lipoproteins and cell membranes. Sterol esterases play a crucial role in the metabolism of lipids by breaking down sterol esters into free sterols and free fatty acids, which can then be used in various biochemical processes.

There are several types of sterol esterases that have been identified, including:

1. Cholesteryl esterase (CE): This enzyme is responsible for hydrolyzing cholesteryl esters in the intestine and liver. It plays a critical role in the absorption and metabolism of dietary cholesterol.
2. Hormone-sensitive lipase (HSL): This enzyme is involved in the hydrolysis of sterol esters in adipose tissue, as well as other lipids such as triacylglycerols. It is regulated by hormones such as insulin and catecholamines.
3. Carboxylesterase (CES): This enzyme is a broad-specificity esterase that can hydrolyze various types of esters, including sterol esters. It is found in many tissues throughout the body.

Sterol esterases are important targets for drug development, as inhibiting these enzymes can have therapeutic effects in a variety of diseases, such as obesity, diabetes, and cardiovascular disease.

Eosinophils are a type of white blood cell that play an important role in the body's immune response. They are produced in the bone marrow and released into the bloodstream, where they can travel to different tissues and organs throughout the body. Eosinophils are characterized by their granules, which contain various proteins and enzymes that are toxic to parasites and can contribute to inflammation.

Eosinophils are typically associated with allergic reactions, asthma, and other inflammatory conditions. They can also be involved in the body's response to certain infections, particularly those caused by parasites such as worms. In some cases, elevated levels of eosinophils in the blood or tissues (a condition called eosinophilia) can indicate an underlying medical condition, such as a parasitic infection, autoimmune disorder, or cancer.

Eosinophils are named for their staining properties - they readily take up eosin dye, which is why they appear pink or red under the microscope. They make up only about 1-6% of circulating white blood cells in healthy individuals, but their numbers can increase significantly in response to certain triggers.

Antiprotozoal agents are a type of medication used to treat protozoal infections, which are infections caused by microscopic single-celled organisms called protozoa. These agents work by either killing the protozoa or inhibiting their growth and reproduction. They can be administered through various routes, including oral, topical, and intravenous, depending on the type of infection and the severity of the illness.

Examples of antiprotozoal agents include:

* Metronidazole, tinidazole, and nitazoxanide for treating infections caused by Giardia lamblia and Entamoeba histolytica.
* Atovaquone, clindamycin, and pyrimethamine-sulfadoxine for treating malaria caused by Plasmodium falciparum or other Plasmodium species.
* Pentamidine and suramin for treating African trypanosomiasis (sleeping sickness) caused by Trypanosoma brucei gambiense or T. b. rhodesiense.
* Nitroimidazoles, such as benznidazole and nifurtimox, for treating Chagas disease caused by Trypanosoma cruzi.
* Sodium stibogluconate and paromomycin for treating leishmaniasis caused by Leishmania species.

Antiprotozoal agents can have side effects, ranging from mild to severe, depending on the drug and the individual patient's response. It is essential to follow the prescribing physician's instructions carefully when taking these medications and report any adverse reactions promptly.

Homeostasis is a fundamental concept in the field of medicine and physiology, referring to the body's ability to maintain a stable internal environment, despite changes in external conditions. It is the process by which biological systems regulate their internal environment to remain in a state of dynamic equilibrium. This is achieved through various feedback mechanisms that involve sensors, control centers, and effectors, working together to detect, interpret, and respond to disturbances in the system.

For example, the body maintains homeostasis through mechanisms such as temperature regulation (through sweating or shivering), fluid balance (through kidney function and thirst), and blood glucose levels (through insulin and glucagon secretion). When homeostasis is disrupted, it can lead to disease or dysfunction in the body.

In summary, homeostasis is the maintenance of a stable internal environment within biological systems, through various regulatory mechanisms that respond to changes in external conditions.

Recombinant fusion proteins are artificially created biomolecules that combine the functional domains or properties of two or more different proteins into a single protein entity. They are generated through recombinant DNA technology, where the genes encoding the desired protein domains are linked together and expressed as a single, chimeric gene in a host organism, such as bacteria, yeast, or mammalian cells.

The resulting fusion protein retains the functional properties of its individual constituent proteins, allowing for novel applications in research, diagnostics, and therapeutics. For instance, recombinant fusion proteins can be designed to enhance protein stability, solubility, or immunogenicity, making them valuable tools for studying protein-protein interactions, developing targeted therapies, or generating vaccines against infectious diseases or cancer.

Examples of recombinant fusion proteins include:

1. Etaglunatide (ABT-523): A soluble Fc fusion protein that combines the heavy chain fragment crystallizable region (Fc) of an immunoglobulin with the extracellular domain of the human interleukin-6 receptor (IL-6R). This fusion protein functions as a decoy receptor, neutralizing IL-6 and its downstream signaling pathways in rheumatoid arthritis.
2. Etanercept (Enbrel): A soluble TNF receptor p75 Fc fusion protein that binds to tumor necrosis factor-alpha (TNF-α) and inhibits its proinflammatory activity, making it a valuable therapeutic option for treating autoimmune diseases like rheumatoid arthritis, ankylosing spondylitis, and psoriasis.
3. Abatacept (Orencia): A fusion protein consisting of the extracellular domain of cytotoxic T-lymphocyte antigen 4 (CTLA-4) linked to the Fc region of an immunoglobulin, which downregulates T-cell activation and proliferation in autoimmune diseases like rheumatoid arthritis.
4. Belimumab (Benlysta): A monoclonal antibody that targets B-lymphocyte stimulator (BLyS) protein, preventing its interaction with the B-cell surface receptor and inhibiting B-cell activation in systemic lupus erythematosus (SLE).
5. Romiplostim (Nplate): A fusion protein consisting of a thrombopoietin receptor agonist peptide linked to an immunoglobulin Fc region, which stimulates platelet production in patients with chronic immune thrombocytopenia (ITP).
6. Darbepoetin alfa (Aranesp): A hyperglycosylated erythropoiesis-stimulating protein that functions as a longer-acting form of recombinant human erythropoietin, used to treat anemia in patients with chronic kidney disease or cancer.
7. Palivizumab (Synagis): A monoclonal antibody directed against the F protein of respiratory syncytial virus (RSV), which prevents RSV infection and is administered prophylactically to high-risk infants during the RSV season.
8. Ranibizumab (Lucentis): A recombinant humanized monoclonal antibody fragment that binds and inhibits vascular endothelial growth factor A (VEGF-A), used in the treatment of age-related macular degeneration, diabetic retinopathy, and other ocular disorders.
9. Cetuximab (Erbitux): A chimeric monoclonal antibody that binds to epidermal growth factor receptor (EGFR), used in the treatment of colorectal cancer and head and neck squamous cell carcinoma.
10. Adalimumab (Humira): A fully humanized monoclonal antibody that targets tumor necrosis factor-alpha (TNF-α), used in the treatment of various inflammatory diseases, including rheumatoid arthritis, psoriasis, and Crohn's disease.
11. Bevacizumab (Avastin): A recombinant humanized monoclonal antibody that binds to VEGF-A, used in the treatment of various cancers, including colorectal, lung, breast, and kidney cancer.
12. Trastuzumab (Herceptin): A humanized monoclonal antibody that targets HER2/neu receptor, used in the treatment of breast cancer.
13. Rituximab (Rituxan): A chimeric monoclonal antibody that binds to CD20 antigen on B cells, used in the treatment of non-Hodgkin's lymphoma and rheumatoid arthritis.
14. Palivizumab (Synagis): A humanized monoclonal antibody that binds to the F protein of respiratory syncytial virus, used in the prevention of respiratory syncytial virus infection in high-risk infants.
15. Infliximab (Remicade): A chimeric monoclonal antibody that targets TNF-α, used in the treatment of various inflammatory diseases, including Crohn's disease, ulcerative colitis, rheumatoid arthritis, and ankylosing spondylitis.
16. Natalizumab (Tysabri): A humanized monoclonal antibody that binds to α4β1 integrin, used in the treatment of multiple sclerosis and Crohn's disease.
17. Adalimumab (Humira): A fully human monoclonal antibody that targets TNF-α, used in the treatment of various inflammatory diseases, including rheumatoid arthritis, psoriatic arthritis, ankylosing spondylitis, Crohn's disease, and ulcerative colitis.
18. Golimumab (Simponi): A fully human monoclonal antibody that targets TNF-α, used in the treatment of rheumatoid arthritis, psoriatic arthritis, ankylosing spondylitis, and ulcerative colitis.
19. Certolizumab pegol (Cimzia): A PEGylated Fab' fragment of a humanized monoclonal antibody that targets TNF-α, used in the treatment of rheumatoid arthritis, psoriatic arthritis, ankylosing spondylitis, and Crohn's disease.
20. Ustekinumab (Stelara): A fully human monoclonal antibody that targets IL-12 and IL-23, used in the treatment of psoriasis, psoriatic arthritis, and Crohn's disease.
21. Secukinumab (Cosentyx): A fully human monoclonal antibody that targets IL-17A, used in the treatment of psoriasis, psoriatic arthritis, and ankylosing spondylitis.
22. Ixekizumab (Taltz): A fully human monoclonal antibody that targets IL-17A, used in the treatment of psoriasis and psoriatic arthritis.
23. Brodalumab (Siliq): A fully human monoclonal antibody that targets IL-17 receptor A, used in the treatment of psoriasis.
24. Sarilumab (Kevzara): A fully human monoclonal antibody that targets the IL-6 receptor, used in the treatment of rheumatoid arthritis.
25. Tocilizumab (Actemra): A humanized monoclonal antibody that targets the IL-6 receptor, used in the treatment of rheumatoid arthritis, systemic juvenile idiopathic arthritis, polyarticular juvenile idiopathic arthritis, giant cell arteritis, and chimeric antigen receptor T-cell-induced cytokine release syndrome.
26. Siltuximab (Sylvant): A chimeric monoclonal antibody that targets IL-6, used in the treatment of multicentric Castleman disease.
27. Satralizumab (Enspryng): A humanized monoclonal antibody that targets IL-6 receptor alpha, used in the treatment of neuromyelitis optica spectrum disorder.
28. Sirukumab (Plivensia): A human monoclonal antibody that targets IL-6, used in the treatment

Esterification is a chemical reaction that involves the conversion of an alcohol and a carboxylic acid into an ester, typically through the removal of a molecule of water. This reaction is often catalyzed by an acid or a base, and it is a key process in organic chemistry. Esters are commonly found in nature and are responsible for the fragrances of many fruits and flowers. They are also important in the production of various industrial and consumer products, including plastics, resins, and perfumes.

Chemokine (C-C motif) ligand 20, also known as CCL20 or EXODUS, is a small signaling protein that belongs to the chemokine family. Chemokines are a group of cytokines, or cell signaling molecules, that play crucial roles in immune responses and inflammation by recruiting various immune cells to the sites of infection or injury.

CCL20 specifically binds to its receptor CCR6 and plays an essential role in attracting immune cells like T lymphocytes (T cells), dendritic cells, and B lymphocytes (B cells) to the site of inflammation. It is produced by various cell types, including epithelial cells, fibroblasts, and immune cells, in response to infection, injury, or other stimuli.

CCL20 has been implicated in several physiological and pathological processes, such as:

1. Homeostatic regulation of immune cell trafficking: CCL20 helps maintain the normal migration and positioning of immune cells in various tissues under steady-state conditions.
2. Inflammatory responses: CCL20 is upregulated during inflammation, contributing to the recruitment of immune cells to the affected area.
3. Autoimmune diseases: Overexpression or dysregulation of CCL20 has been associated with several autoimmune disorders, such as rheumatoid arthritis, psoriasis, and multiple sclerosis.
4. Cancer: CCL20 is involved in tumorigenesis and cancer progression by promoting the recruitment of immune cells that can either support or suppress tumor growth.
5. Infectious diseases: CCL20 plays a role in host defense against various pathogens, including bacteria, viruses, and parasites, by attracting immune cells to the site of infection.

Proto-oncogene proteins, such as c-HCK (hemapoietic cell kinase), are normal cellular proteins that play crucial roles in various cellular processes, including signal transduction, cell cycle regulation, and differentiation. They are involved in the regulation of cell growth and division under physiological conditions.

When proto-oncogenes undergo mutations or aberrant regulation, they can become oncogenes, leading to uncontrolled cell growth and division, which may contribute to cancer development. The c-HCK protein is a non-receptor tyrosine kinase that belongs to the Src family of kinases. It is primarily expressed in hematopoietic cells and plays essential roles in signal transduction pathways involved in cell proliferation, differentiation, and survival.

Mutations or aberrant regulation of c-HCK can lead to its hyperactivation, which may contribute to the development and progression of certain types of leukemias and lymphomas.

AIDS Dementia Complex (ADC) is a neurological disorder that occurs in people with advanced HIV infection or AIDS. It is also known as HIV-associated dementia (HAD) or HIV encephalopathy. ADC is characterized by cognitive impairment, motor dysfunction, and behavioral changes that can significantly affect the individual's daily functioning and quality of life.

The symptoms of AIDS Dementia Complex may include:
- Difficulty with concentration and memory
- Slowness in thinking, processing information, or making decisions
- Changes in mood or personality, such as depression, irritability, or apathy
- Difficulty with coordination, balance, or speech
- Progressive weakness and wasting of muscles
- Difficulty with swallowing or speaking

The exact cause of ADC is not fully understood, but it is believed to be related to the direct effects of HIV on the brain. The virus can infect and damage nerve cells, leading to inflammation and degeneration of brain tissue. Treatment for ADC typically involves antiretroviral therapy (ART) to control HIV replication, as well as medications to manage specific symptoms. In some cases, supportive care such as physical therapy or occupational therapy may also be recommended.

"Mycobacterium" is a genus of gram-positive, aerobic, rod-shaped bacteria that are characterized by their complex cell walls containing large amounts of lipids. This genus includes several species that are significant in human and animal health, most notably Mycobacterium tuberculosis, which causes tuberculosis, and Mycobacterium leprae, which causes leprosy. Other species of Mycobacterium can cause various diseases in humans, including skin and soft tissue infections, lung infections, and disseminated disease in immunocompromised individuals. These bacteria are often resistant to common disinfectants and antibiotics, making them difficult to treat.

I'm sorry for any confusion, but "quartz" is not a medical term. Quartz is a mineral that is one of the most common minerals on Earth. It is found in many different types of rocks and is made up of silicon dioxide (SiO2). It is often used in electronics and watches because of its piezoelectric properties, which means it can generate an electric charge when subjected to pressure or stress.

It does not have any direct relation with medical field, although there are some Quartz based equipment like quartz crystal microbalance (QCM) sensors that are used in medical research for the detection of biomolecules and pathogens.

Disease progression is the worsening or advancement of a medical condition over time. It refers to the natural course of a disease, including its development, the severity of symptoms and complications, and the impact on the patient's overall health and quality of life. Understanding disease progression is important for developing appropriate treatment plans, monitoring response to therapy, and predicting outcomes.

The rate of disease progression can vary widely depending on the type of medical condition, individual patient factors, and the effectiveness of treatment. Some diseases may progress rapidly over a short period of time, while others may progress more slowly over many years. In some cases, disease progression may be slowed or even halted with appropriate medical interventions, while in other cases, the progression may be inevitable and irreversible.

In clinical practice, healthcare providers closely monitor disease progression through regular assessments, imaging studies, and laboratory tests. This information is used to guide treatment decisions and adjust care plans as needed to optimize patient outcomes and improve quality of life.

High Mobility Group Box 1 (HMGB1) protein is a non-histone chromosomal protein that is widely expressed in various cell types, including immune cells and nucleated cells. It plays a crucial role in the maintenance of nucleosome structure and stability, regulation of gene transcription, and DNA replication and repair. HMGB1 can be actively secreted by activated immune cells or passively released from necrotic or damaged cells. Once outside the cell, it functions as a damage-associated molecular pattern (DAMP) molecule that binds to various receptors, such as Toll-like receptors and the receptor for advanced glycation end products (RAGE), on immune cells, leading to the activation of inflammatory responses and the induction of innate and adaptive immunity. HMGB1 has been implicated in various physiological and pathological processes, including inflammation, infection, autoimmunity, cancer, and neurological disorders.

Pathologic neovascularization is the abnormal growth of new blood vessels in previously avascular tissue or excessive growth within existing vasculature, which occurs as a result of hypoxia, inflammation, or angiogenic stimuli. These newly formed vessels are often disorganized, fragile, and lack proper vessel hierarchy, leading to impaired blood flow and increased vascular permeability. Pathologic neovascularization can be observed in various diseases such as cancer, diabetic retinopathy, age-related macular degeneration, and chronic inflammation. This process contributes to disease progression by promoting tumor growth, metastasis, and edema formation, ultimately leading to tissue damage and organ dysfunction.

Visceral leishmaniasis (VL), also known as kala-azar, is a systemic protozoan disease caused by the Leishmania donovani complex. It is the most severe form of leishmaniasis and is characterized by fever, weight loss, anemia, hepatosplenomegaly, and pancytopenia. If left untreated, it can be fatal in over 95% of cases within 2 years of onset of symptoms. It is transmitted to humans through the bite of infected female sandflies (Phlebotomus spp. or Lutzomyia spp.). The parasites enter the skin and are taken up by macrophages, where they transform into amastigotes and spread to internal organs such as the spleen, liver, and bone marrow. Diagnosis is typically made through demonstration of the parasite in tissue samples or through serological tests. Treatment options include antimonial drugs, amphotericin B, miltefosine, and paromomycin. Prevention measures include vector control, early detection and treatment, and protection against sandfly bites.

Platelet-activating factor (PAF) is a potent phospholipid mediator that plays a significant role in various inflammatory and immune responses. It is a powerful lipid signaling molecule released mainly by activated platelets, neutrophils, monocytes, endothelial cells, and other cell types during inflammation or injury.

PAF has a molecular structure consisting of an alkyl chain linked to a glycerol moiety, a phosphate group, and an sn-2 acetyl group. This unique structure allows PAF to bind to its specific G protein-coupled receptor (PAF-R) on the surface of target cells, triggering various intracellular signaling cascades that result in cell activation, degranulation, and aggregation.

The primary functions of PAF include:

1. Platelet activation and aggregation: PAF stimulates platelets to aggregate, release their granules, and activate the coagulation cascade, which can lead to thrombus formation.
2. Neutrophil and monocyte activation: PAF activates these immune cells, leading to increased adhesion, degranulation, and production of reactive oxygen species (ROS) and pro-inflammatory cytokines.
3. Vasodilation and increased vascular permeability: PAF can cause vasodilation by acting on endothelial cells, leading to an increase in blood flow and facilitating the extravasation of immune cells into inflamed tissues.
4. Bronchoconstriction: In the respiratory system, PAF can induce bronchoconstriction and recruitment of inflammatory cells, contributing to asthma symptoms.
5. Neurotransmission modulation: PAF has been implicated in neuroinflammation and may play a role in neuronal excitability, synaptic plasticity, and cognitive functions.

Dysregulated PAF signaling has been associated with several pathological conditions, including atherosclerosis, sepsis, acute respiratory distress syndrome (ARDS), ischemia-reperfusion injury, and neuroinflammatory disorders. Therefore, targeting the PAF pathway may provide therapeutic benefits in these diseases.

The cell nucleus is a membrane-bound organelle found in the eukaryotic cells (cells with a true nucleus). It contains most of the cell's genetic material, organized as DNA molecules in complex with proteins, RNA molecules, and histones to form chromosomes.

The primary function of the cell nucleus is to regulate and control the activities of the cell, including growth, metabolism, protein synthesis, and reproduction. It also plays a crucial role in the process of mitosis (cell division) by separating and protecting the genetic material during this process. The nuclear membrane, or nuclear envelope, surrounding the nucleus is composed of two lipid bilayers with numerous pores that allow for the selective transport of molecules between the nucleoplasm (nucleus interior) and the cytoplasm (cell exterior).

The cell nucleus is a vital structure in eukaryotic cells, and its dysfunction can lead to various diseases, including cancer and genetic disorders.

Cytoplasm is the material within a eukaryotic cell (a cell with a true nucleus) that lies between the nuclear membrane and the cell membrane. It is composed of an aqueous solution called cytosol, in which various organelles such as mitochondria, ribosomes, endoplasmic reticulum, Golgi apparatus, lysosomes, and vacuoles are suspended. Cytoplasm also contains a variety of dissolved nutrients, metabolites, ions, and enzymes that are involved in various cellular processes such as metabolism, signaling, and transport. It is where most of the cell's metabolic activities take place, and it plays a crucial role in maintaining the structure and function of the cell.

"Swine" is a common term used to refer to even-toed ungulates of the family Suidae, including domestic pigs and wild boars. However, in a medical context, "swine" often appears in the phrase "swine flu," which is a strain of influenza virus that typically infects pigs but can also cause illness in humans. The 2009 H1N1 pandemic was caused by a new strain of swine-origin influenza A virus, which was commonly referred to as "swine flu." It's important to note that this virus is not transmitted through eating cooked pork products; it spreads from person to person, mainly through respiratory droplets produced when an infected person coughs or sneezes.

Molecular weight, also known as molecular mass, is the mass of a molecule. It is expressed in units of atomic mass units (amu) or daltons (Da). Molecular weight is calculated by adding up the atomic weights of each atom in a molecule. It is a useful property in chemistry and biology, as it can be used to determine the concentration of a substance in a solution, or to calculate the amount of a substance that will react with another in a chemical reaction.

Trypanosoma cruzi is a protozoan parasite that causes Chagas disease, also known as American trypanosomiasis. It's transmitted to humans and other mammals through the feces of triatomine bugs, often called "kissing bugs." The parasite can also be spread through contaminated food, drink, or from mother to baby during pregnancy or birth.

The life cycle of Trypanosoma cruzi involves two main forms: the infective metacyclic trypomastigote that is found in the bug's feces and the replicative intracellular amastigote that resides within host cells. The metacyclic trypomastigotes enter the host through mucous membranes or skin lesions, where they invade various types of cells and differentiate into amastigotes. These amastigotes multiply by binary fission and then differentiate back into trypomastigotes, which are released into the bloodstream when the host cell ruptures. The circulating trypomastigotes can then infect other cells or be taken up by another triatomine bug during a blood meal, continuing the life cycle.

Clinical manifestations of Chagas disease range from an acute phase with non-specific symptoms like fever, swelling, and fatigue to a chronic phase characterized by cardiac and gastrointestinal complications, which can develop decades after the initial infection. Early detection and treatment of Chagas disease are crucial for preventing long-term health consequences.

Intercellular signaling peptides and proteins are molecules that mediate communication and interaction between different cells in living organisms. They play crucial roles in various biological processes, including cell growth, differentiation, migration, and apoptosis (programmed cell death). These signals can be released into the extracellular space, where they bind to specific receptors on the target cell's surface, triggering intracellular signaling cascades that ultimately lead to a response.

Peptides are short chains of amino acids, while proteins are larger molecules made up of one or more polypeptide chains. Both can function as intercellular signaling molecules by acting as ligands for cell surface receptors or by being cleaved from larger precursor proteins and released into the extracellular space. Examples of intercellular signaling peptides and proteins include growth factors, cytokines, chemokines, hormones, neurotransmitters, and their respective receptors.

These molecules contribute to maintaining homeostasis within an organism by coordinating cellular activities across tissues and organs. Dysregulation of intercellular signaling pathways has been implicated in various diseases, such as cancer, autoimmune disorders, and neurodegenerative conditions. Therefore, understanding the mechanisms underlying intercellular signaling is essential for developing targeted therapies to treat these disorders.

A "cell line, transformed" is a type of cell culture that has undergone a stable genetic alteration, which confers the ability to grow indefinitely in vitro, outside of the organism from which it was derived. These cells have typically been immortalized through exposure to chemical or viral carcinogens, or by introducing specific oncogenes that disrupt normal cell growth regulation pathways.

Transformed cell lines are widely used in scientific research because they offer a consistent and renewable source of biological material for experimentation. They can be used to study various aspects of cell biology, including signal transduction, gene expression, drug discovery, and toxicity testing. However, it is important to note that transformed cells may not always behave identically to their normal counterparts, and results obtained using these cells should be validated in more physiologically relevant systems when possible.

Nitroblue Tetrazolium (NBT) is not a medical term per se, but a chemical compound that is widely used in scientific research and diagnostic tests. It's primarily used as an electron acceptor in various biochemical assays to detect the presence of certain enzymes or reactive oxygen species (ROS).

In a medical context, NBT is often used in the NBT reduction test, which is a diagnostic procedure to identify patients with chronic granulomatous disease (CGD), an inherited immunodeficiency disorder. In this test, white blood cells called phagocytes from the patient's blood sample are incubated with NBT and a stimulus that triggers their respiratory burst, such as bacterial particles. If the phagocytes can produce superoxide radicals during the respiratory burst, these radicals reduce NBT to form a blue-black insoluble formazan precipitate. In CGD patients, who have impaired production of ROS, there is no or significantly reduced formazan formation, indicating an abnormal NBT reduction test result.

Polymyxin B is an antibiotic derived from the bacterium Paenibacillus polymyxa. It belongs to the class of polypeptide antibiotics and has a cyclic structure with a hydrophobic and a hydrophilic region, which allows it to interact with and disrupt the bacterial cell membrane. Polymyxin B is primarily active against gram-negative bacteria, including many multidrug-resistant strains. It is used clinically to treat serious infections caused by these organisms, such as sepsis, pneumonia, and urinary tract infections. However, its use is limited due to potential nephrotoxicity and neurotoxicity.

In the context of medical and biological sciences, a "binding site" refers to a specific location on a protein, molecule, or cell where another molecule can attach or bind. This binding interaction can lead to various functional changes in the original protein or molecule. The other molecule that binds to the binding site is often referred to as a ligand, which can be a small molecule, ion, or even another protein.

The binding between a ligand and its target binding site can be specific and selective, meaning that only certain ligands can bind to particular binding sites with high affinity. This specificity plays a crucial role in various biological processes, such as signal transduction, enzyme catalysis, or drug action.

In the case of drug development, understanding the location and properties of binding sites on target proteins is essential for designing drugs that can selectively bind to these sites and modulate protein function. This knowledge can help create more effective and safer therapeutic options for various diseases.

Complement C3 is a protein that plays a central role in the complement system, which is a part of the immune system that helps to clear pathogens and damaged cells from the body. Complement C3 can be activated through three different pathways: the classical pathway, the lectin pathway, and the alternative pathway. Once activated, it breaks down into two fragments, C3a and C3b.

C3a is an anaphylatoxin that helps to recruit immune cells to the site of infection or injury, while C3b plays a role in opsonization, which is the process of coating pathogens or damaged cells with proteins to make them more recognizable to the immune system. Additionally, C3b can also activate the membrane attack complex (MAC), which forms a pore in the membrane of target cells leading to their lysis or destruction.

In summary, Complement C3 is an important protein in the complement system that helps to identify and eliminate pathogens and damaged cells from the body through various mechanisms.

Sepsis is a life-threatening condition that arises when the body's response to an infection injures its own tissues and organs. It is characterized by a whole-body inflammatory state (systemic inflammation) that can lead to blood clotting issues, tissue damage, and multiple organ failure.

Sepsis happens when an infection you already have triggers a chain reaction throughout your body. Infections that lead to sepsis most often start in the lungs, urinary tract, skin, or gastrointestinal tract.

Sepsis is a medical emergency. If you suspect sepsis, seek immediate medical attention. Early recognition and treatment of sepsis are crucial to improve outcomes. Treatment usually involves antibiotics, intravenous fluids, and may require oxygen, medication to raise blood pressure, and corticosteroids. In severe cases, surgery may be required to clear the infection.

I apologize, but "sheep" is not a term that has a medical definition. It is a common animal with the scientific name Ovis aries. If you have any medical or health-related questions, I would be happy to try and help answer those for you.

Growth Differentiation Factor 15 (GDF15) is a member of the transforming growth factor-β (TGF-β) superfamily of cytokines, which are signaling proteins involved in various biological processes such as cell growth, differentiation, and apoptosis. GDF15 was originally identified as a protein induced during the development of the mouse placenta, but it is now known to be widely expressed in various tissues in response to stress, injury, or disease.

GDF15 has been shown to have both pro- and anti-inflammatory effects, depending on the context. It can inhibit the production of pro-inflammatory cytokines and promote the differentiation of regulatory T cells, which help to dampen immune responses. On the other hand, GDF15 has also been shown to induce the expression of pro-inflammatory genes in certain cell types, suggesting that its effects may be context-dependent.

In terms of its role in growth and differentiation, GDF15 has been implicated in a variety of processes, including the regulation of energy metabolism, appetite control, and tissue repair. For example, GDF15 has been shown to inhibit food intake and promote weight loss in both mice and humans, suggesting that it may play a role in the regulation of body weight. Additionally, GDF15 has been implicated in the development of certain diseases, such as cancer, heart disease, and neurological disorders, although its precise role in these conditions is not yet fully understood.

Overall, GDF15 is a multifunctional cytokine that plays important roles in various biological processes, including inflammation, growth, differentiation, and metabolism. Its precise functions and mechanisms of action are still being elucidated, but it is clear that GDF15 has significant potential as a therapeutic target for a variety of diseases.

Beta-glucans are a type of complex carbohydrate known as polysaccharides, which are found in the cell walls of certain cereals, bacteria, and fungi, including baker's yeast, mushrooms, and algae. They consist of long chains of glucose molecules linked together by beta-glycosidic bonds.

Beta-glucans have been studied for their potential health benefits, such as boosting the immune system, reducing cholesterol levels, and improving gut health. They are believed to work by interacting with immune cells, such as macrophages and neutrophils, and enhancing their ability to recognize and destroy foreign invaders like bacteria, viruses, and tumor cells.

Beta-glucans are available in supplement form and are also found in various functional foods and beverages, such as baked goods, cereals, and sports drinks. However, it is important to note that the effectiveness of beta-glucans for these health benefits may vary depending on the source, dose, and individual's health status. Therefore, it is recommended to consult with a healthcare professional before taking any dietary supplements or making significant changes to your diet.

Immunoglobulin Fc fragments are the crystallizable fragment of an antibody that is responsible for effector functions such as engagement with Fc receptors on immune cells, activation of the complement system, and neutralization of toxins. The Fc region is located at the tail end of the Y-shaped immunoglobulin molecule, and it is made up of constant regions of the heavy chains of the antibody.

When an antibody binds to its target antigen, the Fc region can interact with other proteins in the immune system, leading to a variety of responses such as phagocytosis, antibody-dependent cellular cytotoxicity (ADCC), and complement activation. These effector functions help to eliminate pathogens and infected cells from the body.

Immunoglobulin Fc fragments can be produced artificially through enzymatic digestion of intact antibodies, resulting in a fragment that retains the ability to interact with Fc receptors and other proteins involved in immune responses. These fragments have potential therapeutic applications in a variety of diseases, including autoimmune disorders, inflammatory conditions, and cancer.

Adaptor proteins are a type of protein that play a crucial role in intracellular signaling pathways by serving as a link between different components of the signaling complex. Specifically, "signal transducing adaptor proteins" refer to those adaptor proteins that are involved in signal transduction processes, where they help to transmit signals from the cell surface receptors to various intracellular effectors. These proteins typically contain modular domains that allow them to interact with multiple partners, thereby facilitating the formation of large signaling complexes and enabling the integration of signals from different pathways.

Signal transducing adaptor proteins can be classified into several families based on their structural features, including the Src homology 2 (SH2) domain, the Src homology 3 (SH3) domain, and the phosphotyrosine-binding (PTB) domain. These domains enable the adaptor proteins to recognize and bind to specific motifs on other signaling molecules, such as receptor tyrosine kinases, G protein-coupled receptors, and cytokine receptors.

One well-known example of a signal transducing adaptor protein is the growth factor receptor-bound protein 2 (Grb2), which contains an SH2 domain that binds to phosphotyrosine residues on activated receptor tyrosine kinases. Grb2 also contains an SH3 domain that interacts with proline-rich motifs on other signaling proteins, such as the guanine nucleotide exchange factor SOS. This interaction facilitates the activation of the Ras small GTPase and downstream signaling pathways involved in cell growth, differentiation, and survival.

Overall, signal transducing adaptor proteins play a critical role in regulating various cellular processes by modulating intracellular signaling pathways in response to extracellular stimuli. Dysregulation of these proteins has been implicated in various diseases, including cancer and inflammatory disorders.

HIV (Human Immunodeficiency Virus) infection is a viral illness that progressively attacks and weakens the immune system, making individuals more susceptible to other infections and diseases. The virus primarily infects CD4+ T cells, a type of white blood cell essential for fighting off infections. Over time, as the number of these immune cells declines, the body becomes increasingly vulnerable to opportunistic infections and cancers.

HIV infection has three stages:

1. Acute HIV infection: This is the initial stage that occurs within 2-4 weeks after exposure to the virus. During this period, individuals may experience flu-like symptoms such as fever, fatigue, rash, swollen glands, and muscle aches. The virus replicates rapidly, and the viral load in the body is very high.
2. Chronic HIV infection (Clinical latency): This stage follows the acute infection and can last several years if left untreated. Although individuals may not show any symptoms during this phase, the virus continues to replicate at low levels, and the immune system gradually weakens. The viral load remains relatively stable, but the number of CD4+ T cells declines over time.
3. AIDS (Acquired Immunodeficiency Syndrome): This is the most advanced stage of HIV infection, characterized by a severely damaged immune system and numerous opportunistic infections or cancers. At this stage, the CD4+ T cell count drops below 200 cells/mm3 of blood.

It's important to note that with proper antiretroviral therapy (ART), individuals with HIV infection can effectively manage the virus, maintain a healthy immune system, and significantly reduce the risk of transmission to others. Early diagnosis and treatment are crucial for improving long-term health outcomes and reducing the spread of HIV.

RNA (Ribonucleic Acid) is a single-stranded, linear polymer of ribonucleotides. It is a nucleic acid present in the cells of all living organisms and some viruses. RNAs play crucial roles in various biological processes such as protein synthesis, gene regulation, and cellular signaling. There are several types of RNA including messenger RNA (mRNA), ribosomal RNA (rRNA), transfer RNA (tRNA), small nuclear RNA (snRNA), microRNA (miRNA), and long non-coding RNA (lncRNA). These RNAs differ in their structure, function, and location within the cell.

The brain is the central organ of the nervous system, responsible for receiving and processing sensory information, regulating vital functions, and controlling behavior, movement, and cognition. It is divided into several distinct regions, each with specific functions:

1. Cerebrum: The largest part of the brain, responsible for higher cognitive functions such as thinking, learning, memory, language, and perception. It is divided into two hemispheres, each controlling the opposite side of the body.
2. Cerebellum: Located at the back of the brain, it is responsible for coordinating muscle movements, maintaining balance, and fine-tuning motor skills.
3. Brainstem: Connects the cerebrum and cerebellum to the spinal cord, controlling vital functions such as breathing, heart rate, and blood pressure. It also serves as a relay center for sensory information and motor commands between the brain and the rest of the body.
4. Diencephalon: A region that includes the thalamus (a major sensory relay station) and hypothalamus (regulates hormones, temperature, hunger, thirst, and sleep).
5. Limbic system: A group of structures involved in emotional processing, memory formation, and motivation, including the hippocampus, amygdala, and cingulate gyrus.

The brain is composed of billions of interconnected neurons that communicate through electrical and chemical signals. It is protected by the skull and surrounded by three layers of membranes called meninges, as well as cerebrospinal fluid that provides cushioning and nutrients.

Non-steroidal anti-inflammatory agents (NSAIDs) are a class of medications that reduce pain, inflammation, and fever. They work by inhibiting the activity of cyclooxygenase (COX) enzymes, which are involved in the production of prostaglandins, chemicals that contribute to inflammation and cause blood vessels to dilate and become more permeable, leading to symptoms such as pain, redness, warmth, and swelling.

NSAIDs are commonly used to treat a variety of conditions, including arthritis, muscle strains and sprains, menstrual cramps, headaches, and fever. Some examples of NSAIDs include aspirin, ibuprofen, naproxen, and celecoxib.

While NSAIDs are generally safe and effective when used as directed, they can have side effects, particularly when taken in large doses or for long periods of time. Common side effects include stomach ulcers, gastrointestinal bleeding, and increased risk of heart attack and stroke. It is important to follow the recommended dosage and consult with a healthcare provider if you have any concerns about using NSAIDs.

Histiocytes are a type of immune cell that are part of the mononuclear phagocyte system. They originate from monocytes, which are derived from hematopoietic stem cells in the bone marrow. Histiocytes play an important role in the immune system by engulfing and destroying foreign substances, such as bacteria and viruses, as well as removing dead cells and other debris from the body. They can be found in various tissues throughout the body, including the skin, lymph nodes, spleen, and liver.

Histiocytes include several different types of cells, such as macrophages, dendritic cells, and Langerhans cells. These cells have different functions but all play a role in the immune response. For example, macrophages are involved in inflammation and tissue repair, while dendritic cells are important for presenting antigens to T cells and initiating an immune response.

Abnormal accumulations or dysfunction of histiocytes can lead to various diseases, such as histiocytosis, which is a group of disorders characterized by the abnormal proliferation and accumulation of histiocytes in various tissues.

A plant extract is a preparation containing chemical constituents that have been extracted from a plant using a solvent. The resulting extract may contain a single compound or a mixture of several compounds, depending on the extraction process and the specific plant material used. These extracts are often used in various industries including pharmaceuticals, nutraceuticals, cosmetics, and food and beverage, due to their potential therapeutic or beneficial properties. The composition of plant extracts can vary widely, and it is important to ensure their quality, safety, and efficacy before use in any application.

Hydrogen-ion concentration, also known as pH, is a measure of the acidity or basicity of a solution. It is defined as the negative logarithm (to the base 10) of the hydrogen ion activity in a solution. The standard unit of measurement is the pH unit. A pH of 7 is neutral, less than 7 is acidic, and greater than 7 is basic.

In medical terms, hydrogen-ion concentration is important for maintaining homeostasis within the body. For example, in the stomach, a high hydrogen-ion concentration (low pH) is necessary for the digestion of food. However, in other parts of the body such as blood, a high hydrogen-ion concentration can be harmful and lead to acidosis. Conversely, a low hydrogen-ion concentration (high pH) in the blood can lead to alkalosis. Both acidosis and alkalosis can have serious consequences on various organ systems if not corrected.

JNK (c-Jun N-terminal kinase) Mitogen-Activated Protein Kinases are a subgroup of the Ser/Thr protein kinases that are activated by stress stimuli and play important roles in various cellular processes, including inflammation, apoptosis, and differentiation. They are involved in the regulation of gene expression through phosphorylation of transcription factors such as c-Jun. JNKs are activated by a variety of upstream kinases, including MAP2Ks (MKK4/SEK1 and MKK7), which are in turn activated by MAP3Ks (such as ASK1, MEKK1, MLKs, and TAK1). JNK signaling pathways have been implicated in various diseases, including cancer, neurodegenerative disorders, and inflammatory diseases.

A kidney glomerulus is a functional unit in the nephron of the kidney. It is a tuft of capillaries enclosed within a structure called Bowman's capsule, which filters waste and excess fluids from the blood. The glomerulus receives blood from an afferent arteriole and drains into an efferent arteriole.

The process of filtration in the glomerulus is called ultrafiltration, where the pressure within the glomerular capillaries drives plasma fluid and small molecules (such as ions, glucose, amino acids, and waste products) through the filtration membrane into the Bowman's space. Larger molecules, like proteins and blood cells, are retained in the blood due to their larger size. The filtrate then continues down the nephron for further processing, eventually forming urine.

Mast cells are a type of white blood cell that are found in connective tissues throughout the body, including the skin, respiratory tract, and gastrointestinal tract. They play an important role in the immune system and help to defend the body against pathogens by releasing chemicals such as histamine, heparin, and leukotrienes, which help to attract other immune cells to the site of infection or injury. Mast cells also play a role in allergic reactions, as they release histamine and other chemicals in response to exposure to an allergen, leading to symptoms such as itching, swelling, and redness. They are derived from hematopoietic stem cells in the bone marrow and mature in the tissues where they reside.

Mannans are a type of complex carbohydrate, specifically a heteropolysaccharide, that are found in the cell walls of certain plants, algae, and fungi. They consist of chains of mannose sugars linked together, often with other sugar molecules such as glucose or galactose.

Mannans have various biological functions, including serving as a source of energy for microorganisms that can break them down. In some cases, mannans can also play a role in the immune response and are used as a component of vaccines to stimulate an immune response.

In the context of medicine, mannans may be relevant in certain conditions such as gut dysbiosis or allergic reactions to foods containing mannans. Additionally, some research has explored the potential use of mannans as a delivery vehicle for drugs or other therapeutic agents.

Cytochalasin D is a toxin produced by certain fungi that inhibits the polymerization and elongation of actin filaments, which are crucial components of the cytoskeleton in cells. This results in the disruption of various cellular processes such as cell division, motility, and shape maintenance. It is often used in research to study actin dynamics and cellular structure.

Lipoprotein lipase (LPL) is an enzyme that plays a crucial role in the metabolism of lipids. It is responsible for breaking down triglycerides, which are the main constituent of dietary fats and chylomicrons, into fatty acids and glycerol. These products are then taken up by cells for energy production or storage.

LPL is synthesized in various tissues, including muscle and fat, where it is attached to the inner lining of blood vessels (endothelium). The enzyme is activated when it comes into contact with lipoprotein particles, such as chylomicrons and very-low-density lipoproteins (VLDL), which transport triglycerides in the bloodstream.

Deficiencies or mutations in LPL can lead to various metabolic disorders, including hypertriglyceridemia, a condition characterized by high levels of triglycerides in the blood. Conversely, overexpression of LPL has been associated with increased risk of atherosclerosis due to excessive uptake of fatty acids by macrophages and their conversion into foam cells, which contribute to plaque formation in the arteries.

Mycobacterium avium Complex (MAC) is a group of slow-growing mycobacteria that includes Mycobacterium avium and Mycobacterium intracellulare. These bacteria are commonly found in water, soil, and dust, and can cause pulmonary disease, lymphadenitis, and disseminated infection, particularly in individuals with compromised immune systems, such as those with HIV/AIDS. The infection caused by MAC is often chronic and difficult to eradicate, requiring long-term antibiotic therapy.

Solubility is a fundamental concept in pharmaceutical sciences and medicine, which refers to the maximum amount of a substance (solute) that can be dissolved in a given quantity of solvent (usually water) at a specific temperature and pressure. Solubility is typically expressed as mass of solute per volume or mass of solvent (e.g., grams per liter, milligrams per milliliter). The process of dissolving a solute in a solvent results in a homogeneous solution where the solute particles are dispersed uniformly throughout the solvent.

Understanding the solubility of drugs is crucial for their formulation, administration, and therapeutic effectiveness. Drugs with low solubility may not dissolve sufficiently to produce the desired pharmacological effect, while those with high solubility might lead to rapid absorption and short duration of action. Therefore, optimizing drug solubility through various techniques like particle size reduction, salt formation, or solubilization is an essential aspect of drug development and delivery.

The thymus gland is an essential organ of the immune system, located in the upper chest, behind the sternum and surrounding the heart. It's primarily active until puberty and begins to shrink in size and activity thereafter. The main function of the thymus gland is the production and maturation of T-lymphocytes (T-cells), which are crucial for cell-mediated immunity, helping to protect the body from infection and cancer.

The thymus gland provides a protected environment where immune cells called pre-T cells develop into mature T cells. During this process, they learn to recognize and respond appropriately to foreign substances while remaining tolerant to self-tissues, which is crucial for preventing autoimmune diseases.

Additionally, the thymus gland produces hormones like thymosin that regulate immune cell activities and contribute to the overall immune response.

"Mycobacterium smegmatis" is a species of fast-growing, non-tuberculous mycobacteria (NTM). It is commonly found in the environment, including soil and water. This bacterium is known for its ability to form resistant colonies called biofilms. While it does not typically cause disease in humans, it can contaminate medical equipment and samples, potentially leading to misdiagnosis or infection. In rare cases, it has been associated with skin and soft tissue infections. It is often used in research as a model organism for studying mycobacterial biology and drug resistance due to its relatively harmless nature and rapid growth rate.

Fibronectin is a high molecular weight glycoprotein that is found in many tissues and body fluids, including plasma, connective tissue, and the extracellular matrix. It is composed of two similar subunits that are held together by disulfide bonds. Fibronectin plays an important role in cell adhesion, migration, and differentiation by binding to various cell surface receptors, such as integrins, and other extracellular matrix components, such as collagen and heparan sulfate proteoglycans.

Fibronectin has several isoforms that are produced by alternative splicing of a single gene transcript. These isoforms differ in their biological activities and can be found in different tissues and developmental stages. Fibronectin is involved in various physiological processes, such as wound healing, tissue repair, and embryonic development, and has been implicated in several pathological conditions, including fibrosis, tumor metastasis, and thrombosis.

Endosomes are membrane-bound compartments within eukaryotic cells that play a critical role in intracellular trafficking and sorting of various cargoes, including proteins and lipids. They are formed by the invagination of the plasma membrane during endocytosis, resulting in the internalization of extracellular material and cell surface receptors.

Endosomes can be classified into early endosomes, late endosomes, and recycling endosomes based on their morphology, molecular markers, and functional properties. Early endosomes are the initial sorting stations for internalized cargoes, where they undergo sorting and processing before being directed to their final destinations. Late endosomes are more acidic compartments that mature from early endosomes and are responsible for the transport of cargoes to lysosomes for degradation.

Recycling endosomes, on the other hand, are involved in the recycling of internalized cargoes back to the plasma membrane or to other cellular compartments. Endosomal sorting and trafficking are regulated by a complex network of molecular interactions involving various proteins, lipids, and intracellular signaling pathways.

Defects in endosomal function have been implicated in various human diseases, including neurodegenerative disorders, developmental abnormalities, and cancer. Therefore, understanding the mechanisms underlying endosomal trafficking and sorting is of great importance for developing therapeutic strategies to treat these conditions.

Wallerian degeneration is a process that occurs following damage to the axons of neurons (nerve cells). After an axon is severed or traumatically injured, it undergoes a series of changes including fragmentation and removal of the distal segment of the axon, which is the part that is separated from the cell body. This process is named after Augustus Waller, who first described it in 1850.

The degenerative changes in the distal axon are characterized by the breakdown of the axonal cytoskeleton, the loss of myelin sheath (the fatty insulating material that surrounds and protects the axon), and the infiltration of macrophages to clear away the debris. These events lead to the degeneration of the distal axon segment, which is necessary for successful regeneration of the injured nerve.

Wallerian degeneration is a crucial process in the nervous system's response to injury, as it enables the regrowth of axons and the reestablishment of connections between neurons. However, if the regenerative capacity of the neuron is insufficient or the environment is not conducive to growth, functional recovery may be impaired, leading to long-term neurological deficits.

The immune system is a complex network of cells, tissues, and organs that work together to defend the body against harmful invaders. It recognizes and responds to threats such as bacteria, viruses, parasites, fungi, and damaged or abnormal cells, including cancer cells. The immune system has two main components: the innate immune system, which provides a general defense against all types of threats, and the adaptive immune system, which mounts specific responses to particular threats.

The innate immune system includes physical barriers like the skin and mucous membranes, chemical barriers such as stomach acid and enzymes in tears and saliva, and cellular defenses like phagocytes (white blood cells that engulf and destroy invaders) and natural killer cells (which recognize and destroy virus-infected or cancerous cells).

The adaptive immune system is more specific and takes longer to develop a response but has the advantage of "remembering" previous encounters with specific threats. This allows it to mount a faster and stronger response upon subsequent exposures, providing immunity to certain diseases. The adaptive immune system includes T cells (which help coordinate the immune response) and B cells (which produce antibodies that neutralize or destroy invaders).

Overall, the immune system is essential for maintaining health and preventing disease. Dysfunction of the immune system can lead to a variety of disorders, including autoimmune diseases, immunodeficiencies, and allergies.

Beta-N-Acetylhexosaminidases are a group of enzymes that play a role in the breakdown and recycling of complex carbohydrates in the body. Specifically, they help to break down gangliosides, which are a type of molecule found in cell membranes.

There are several different isoforms of beta-N-Acetylhexosaminidases, including A, B, and S. These isoforms are formed by different combinations of subunits, which can affect their activity and substrate specificity.

Mutations in the genes that encode for these enzymes can lead to a variety of genetic disorders, including Tay-Sachs disease and Sandhoff disease. These conditions are characterized by an accumulation of gangliosides in the brain, which can cause progressive neurological deterioration and death.

Treatment for these conditions typically involves managing symptoms and providing supportive care, as there is currently no cure. Enzyme replacement therapy has been explored as a potential treatment option, but its effectiveness varies depending on the specific disorder and the age of the patient.

I'm happy to help! However, I believe there may be a slight mistake in your question. The abbreviation "fms" does not have a clear or established meaning in the context of genetics or molecular biology. It is possible that you may have meant to ask about "FMS-like tyrosine kinase 3 (FLT3)," which is a gene that encodes a type of protein called a receptor tyrosine kinase.

FLT3 plays an important role in the development and function of blood cells, particularly hematopoietic stem cells and progenitor cells. Mutations in the FLT3 gene have been associated with various types of leukemia and other blood disorders. These mutations can lead to increased activity of the FLT3 protein, which can promote the growth and survival of cancerous cells.

Therefore, if you were asking about the definition of "genes fms," I would interpret that as a request for information about the FLT3 gene and its role in genetics and molecular biology.

Interleukin-1 Receptor-Associated Kinases (IRAKs) are a group of serine/threonine protein kinases that play a crucial role in the signaling pathways of Toll-like receptors (TLRs) and Interleukin-1 receptors (IL-1Rs). These receptors are involved in the recognition and response to various pathogen-associated molecular patterns (PAMPs) and damage-associated molecular patterns (DAMPs), which are essential for the activation of innate immune responses.

There are four known members of the IRAK family, namely IRAK1, IRAK2, IRAK3 (also known as IRAK-M), and IRAK4. Among these, IRAK4 is an upstream kinase that gets recruited to the receptor complex upon IL-1R or TLR activation. Once recruited, IRAK4 phosphorylates and activates IRAK1 and IRAK2, which in turn recruit additional signaling proteins leading to the activation of various transcription factors such as NF-κB and AP-1. These transcription factors regulate the expression of genes involved in inflammation, immune response, and cell survival.

IRAK3, on the other hand, is a negative regulator of TLR and IL-1R signaling. It lacks kinase activity and inhibits IRAK1 and IRAK4 activation, thereby dampening the immune response and preventing excessive inflammation. Dysregulation of IRAKs has been implicated in various inflammatory diseases, making them attractive targets for drug development.

CD47 is a cell surface protein that acts as a type of "marker" on certain cells in the body, including red blood cells and immune cells. It is sometimes referred to as an "antigen" because it can be recognized by other proteins called receptors, which can trigger various responses in the body.

CD47 plays a role in regulating the immune response and protecting healthy cells from being attacked by the immune system. It does this by binding to a receptor called SIRPα on certain immune cells, such as macrophages and dendritic cells. This interaction sends a "don't eat me" signal that helps prevent the immune cells from attacking and destroying the CD47-expressing cells.

CD47 has been studied in the context of various diseases, including cancer, because some cancer cells may overexpress CD47 as a way to evade the immune system. Inhibiting the interaction between CD47 and SIRPα has emerged as a potential strategy for enhancing the body's ability to fight off cancer cells.

CD40 is a type of protein known as a tumor necrosis factor receptor that is found on the surface of various cells in the body, including B cells, dendritic cells, and activated T cells. It plays an important role in the immune system by interacting with another protein called CD154 (also known as CD40 ligand) to activate immune responses.

CD40 antigens are molecules that can stimulate an immune response when introduced into the body because they are recognized as foreign substances by the immune system. They may be used in vaccines or other immunotherapies to induce an immune response against specific targets, such as cancer cells or infectious agents.

CD40 antigens can also be found on some types of tumor cells, and activating CD40 with CD154 has been shown to enhance the anti-tumor immune response in preclinical models. Therefore, CD40 agonists are being investigated as potential cancer therapies.

In summary, CD40 antigens are proteins that can stimulate an immune response and are involved in activating immune cells. They have potential applications in vaccines, immunotherapies, and cancer treatments.

CHO cells, or Chinese Hamster Ovary cells, are a type of immortalized cell line that are commonly used in scientific research and biotechnology. They were originally derived from the ovaries of a female Chinese hamster (Cricetulus griseus) in the 1950s.

CHO cells have several characteristics that make them useful for laboratory experiments. They can grow and divide indefinitely under appropriate conditions, which allows researchers to culture large quantities of them for study. Additionally, CHO cells are capable of expressing high levels of recombinant proteins, making them a popular choice for the production of therapeutic drugs, vaccines, and other biologics.

In particular, CHO cells have become a workhorse in the field of biotherapeutics, with many approved monoclonal antibody-based therapies being produced using these cells. The ability to genetically modify CHO cells through various methods has further expanded their utility in research and industrial applications.

It is important to note that while CHO cells are widely used in scientific research, they may not always accurately represent human cell behavior or respond to drugs and other compounds in the same way as human cells do. Therefore, results obtained using CHO cells should be validated in more relevant systems when possible.

Endothelial cells are the type of cells that line the inner surface of blood vessels, lymphatic vessels, and heart chambers. They play a crucial role in maintaining vascular homeostasis by controlling vasomotor tone, coagulation, platelet activation, and inflammation. Endothelial cells also regulate the transport of molecules between the blood and surrounding tissues, and contribute to the maintenance of the structural integrity of the vasculature. They are flat, elongated cells with a unique morphology that allows them to form a continuous, nonthrombogenic lining inside the vessels. Endothelial cells can be isolated from various tissues and cultured in vitro for research purposes.

Arachidonate 12-lipoxygenase (also known as ALOX12 or 12S-lipoxygenase) is an enzyme that catalyzes the conversion of arachidonic acid to 12(S)-hydroperoxyeicosatetraenoic acid (12(S)-HPETE). This reaction is part of the lipoxygenase pathway, which contributes to the biosynthesis of eicosanoids, a group of signaling molecules that play important roles in inflammation and immune response.

The enzyme's function includes introducing molecular oxygen into arachidonic acid at position 12, creating a hydroperoxide group. The product, 12(S)-HPETE, can be further metabolized to various eicosanoids, such as 12-hydroxyeicosatetraenoic acid (12-HETE) and lipoxin A4, which have diverse biological activities in the body.

Arachidonate 12-lipoxygenase is expressed in various tissues, including the vascular endothelium, platelets, and immune cells like monocytes and macrophages. Its activity can contribute to the development of certain diseases, such as atherosclerosis, cancer, and inflammatory disorders. Therefore, inhibiting this enzyme has been considered as a potential therapeutic strategy for treating these conditions.

Monocyte chemoattractant proteins (MCPs) are a group of chemokines, which are small signaling proteins that attract immune cells to sites of infection or inflammation. Specifically, MCPs are responsible for recruiting monocytes and other immune cells to areas of tissue damage or infection.

There are several subtypes of MCPs, including MCP-1 (CCL2), MCP-2 (CCL8), MCP-3 (CCL7), and MCP-4 (CCL13). These proteins bind to specific receptors on the surface of monocytes and other immune cells, triggering a series of intracellular signaling events that result in cell migration towards the site of injury or infection.

MCPs play an important role in the pathogenesis of various inflammatory diseases, such as atherosclerosis, rheumatoid arthritis, and cancer. For example, elevated levels of MCP-1 have been associated with increased monocyte recruitment to the arterial wall, leading to the formation of plaques that can cause heart attacks and strokes. Similarly, high levels of MCPs have been found in the synovial fluid of patients with rheumatoid arthritis, contributing to joint inflammation and damage.

Overall, Monocyte chemoattractant proteins are crucial components of the immune system's response to injury and infection, but their dysregulation can contribute to the development of various diseases.

Acute Monocytic Leukemia (AML-M5) is a subtype of acute myeloid leukemia (AML), which is a type of cancer affecting the blood and bone marrow. In AML-M5, there is an overproduction of abnormal monocytes, a type of white blood cell that normally helps fight infection and is involved in the body's immune response. These abnormal monocytes accumulate in the bone marrow and interfere with the production of normal blood cells, leading to symptoms such as fatigue, frequent infections, and easy bruising or bleeding. The disease progresses rapidly without treatment, making it crucial to begin therapy as soon as possible after diagnosis.

In situ nick-end labeling (ISEL, also known as TUNEL) is a technique used in pathology and molecular biology to detect DNA fragmentation, which is a characteristic of apoptotic cells (cells undergoing programmed cell death). The method involves labeling the 3'-hydroxyl termini of double or single stranded DNA breaks in situ (within tissue sections or individual cells) using modified nucleotides that are coupled to a detectable marker, such as a fluorophore or an enzyme. This technique allows for the direct visualization and quantification of apoptotic cells within complex tissues or cell populations.

Mycobacterium infections are a group of infectious diseases caused by various species of the Mycobacterium genus, including but not limited to M. tuberculosis (which causes tuberculosis), M. avium complex (which causes pulmonary and disseminated disease, particularly in immunocompromised individuals), M. leprae (which causes leprosy), and M. ulcerans (which causes Buruli ulcer). These bacteria are known for their ability to resist destruction by normal immune responses and many disinfectants due to the presence of a waxy mycolic acid layer in their cell walls.

Infection typically occurs through inhalation, ingestion, or direct contact with contaminated materials. The severity and manifestations of the disease can vary widely depending on the specific Mycobacterium species involved, the route of infection, and the host's immune status. Symptoms may include cough, fever, night sweats, weight loss, fatigue, skin lesions, or lymphadenitis. Diagnosis often requires specialized laboratory tests, such as culture or PCR-based methods, to identify the specific Mycobacterium species involved. Treatment typically involves a combination of antibiotics and may require long-term therapy.

STAT3 (Signal Transducer and Activator of Transcription 3) is a transcription factor protein that plays a crucial role in signal transduction and gene regulation. It is activated through phosphorylation by various cytokines and growth factors, which leads to its dimerization, nuclear translocation, and binding to specific DNA sequences. Once bound to the DNA, STAT3 regulates the expression of target genes involved in various cellular processes such as proliferation, differentiation, survival, and angiogenesis. Dysregulation of STAT3 has been implicated in several diseases, including cancer, autoimmune disorders, and inflammatory conditions.

Rosette formation is a term used in pathology and histology, which refers to the circular arrangement of cells or structures around a central point, creating a pattern that resembles a rose flower. This phenomenon can be observed in various tissues and diseases. For example, in the context of cancer, rosette formation may be seen in certain types of tumors, such as medulloblastomas or retinoblastomas, where cancer cells cluster around blood vessels or form distinctive arrangements that are characteristic of these malignancies. In some cases, rosette formation can provide valuable clues for the diagnosis and classification of neoplasms. However, it is essential to consider other histological features and clinical context when interpreting rosette formation in diagnostic pathology.

Cell polarity refers to the asymmetric distribution of membrane components, cytoskeleton, and organelles in a cell. This asymmetry is crucial for various cellular functions such as directed transport, cell division, and signal transduction. The plasma membrane of polarized cells exhibits distinct domains with unique protein and lipid compositions that define apical, basal, and lateral surfaces of the cell.

In epithelial cells, for example, the apical surface faces the lumen or external environment, while the basolateral surface interacts with other cells or the extracellular matrix. The establishment and maintenance of cell polarity are regulated by various factors including protein complexes, lipids, and small GTPases. Loss of cell polarity has been implicated in several diseases, including cancer and neurological disorders.

Salmonella is a genus of rod-shaped, Gram-negative bacteria that are facultative anaerobes and are motile due to peritrichous flagella. They are non-spore forming and often have a single polar flagellum when grown in certain conditions. Salmonella species are important pathogens in humans and other animals, causing foodborne illnesses known as salmonellosis.

Salmonella can be found in the intestinal tracts of humans, birds, reptiles, and mammals. They can contaminate various foods, including meat, poultry, eggs, dairy products, and fresh produce. The bacteria can survive and multiply in a wide range of temperatures and environments, making them challenging to control completely.

Salmonella infection typically leads to gastroenteritis, characterized by symptoms such as diarrhea, abdominal cramps, fever, and vomiting. In some cases, the infection may spread beyond the intestines, leading to more severe complications like bacteremia (bacterial infection of the blood) or focal infections in various organs.

There are two main species of Salmonella: S. enterica and S. bongori. S. enterica is further divided into six subspecies and numerous serovars, with over 2,500 distinct serotypes identified to date. Some well-known Salmonella serovars include S. Typhi (causes typhoid fever), S. Paratyphi A, B, and C (cause paratyphoid fever), and S. Enteritidis and S. Typhimurium (common causes of foodborne salmonellosis).

Chemokine (C-C motif) ligand 7 (CCL7), also known as monocyte chemotactic protein 3 (MCP-3), is a small signaling protein that belongs to the CC-chemokine family. Chemokines are a group of cytokines, or cell signaling molecules, that play crucial roles in immune responses and inflammation by recruiting various immune cells to the sites of infection or injury.

CCL7 is produced by different types of cells, including monocytes, macrophages, fibroblasts, endothelial cells, and certain tumor cells. It exerts its functions by binding to specific chemokine receptors found on the surface of target cells, primarily CCR1, CCR2, and CCR3. The primary role of CCL7 is to attract monocytes, memory T cells, and dendritic cells to the site of inflammation or injury, thereby contributing to the initiation and progression of immune responses.

CCL7 has been implicated in several pathological conditions, such as atherosclerosis, rheumatoid arthritis, cancer, and HIV infection. Its expression is often upregulated during these conditions, leading to excessive recruitment of immune cells, which can result in tissue damage and further exacerbate the disease process. Understanding the role of CCL7 in various diseases may provide insights into developing novel therapeutic strategies for their treatment.

Toll-like receptor 9 (TLR9) is a type of protein belonging to the family of Toll-like receptors, which play a crucial role in the innate immune system. TLR9 is primarily expressed on the endosomal membranes of various immune cells, including dendritic cells, B cells, and macrophages. It recognizes specific molecular patterns, particularly unmethylated CpG DNA motifs, which are commonly found in bacterial and viral genomes but are underrepresented in vertebrate DNA.

Upon recognition and binding to its ligands, TLR9 initiates a signaling cascade that activates various transcription factors, such as NF-κB and IRF7, leading to the production of proinflammatory cytokines, type I interferons, and the activation of adaptive immune responses. This process is essential for the clearance of pathogens and the development of immunity against them. Dysregulation of TLR9 signaling has been implicated in several autoimmune diseases and chronic inflammatory conditions.

Transforming Growth Factor-beta 1 (TGF-β1) is a cytokine that belongs to the TGF-β superfamily. It is a multifunctional protein involved in various cellular processes, including cell growth, differentiation, apoptosis, and extracellular matrix production. TGF-β1 plays crucial roles in embryonic development, tissue homeostasis, and repair, as well as in pathological conditions such as fibrosis and cancer. It signals through a heteromeric complex of type I and type II serine/threonine kinase receptors, leading to the activation of intracellular signaling pathways, primarily the Smad-dependent pathway. TGF-β1 has context-dependent functions, acting as a tumor suppressor in normal and early-stage cancer cells but promoting tumor progression and metastasis in advanced cancers.

'Bacillus anthracis' is the scientific name for the bacterium that causes anthrax, a serious and potentially fatal infectious disease. This gram-positive, spore-forming rod-shaped bacterium can be found in soil and commonly affects animals such as sheep, goats, and cattle. Anthrax can manifest in several forms, including cutaneous (skin), gastrointestinal, and inhalation anthrax, depending on the route of infection.

The spores of Bacillus anthracis are highly resistant to environmental conditions and can survive for years, making them a potential agent for bioterrorism or biowarfare. When inhaled, ingested, or introduced through breaks in the skin, these spores can germinate into vegetative bacteria that produce potent exotoxins responsible for anthrax symptoms and complications.

It is essential to distinguish Bacillus anthracis from other Bacillus species due to its public health significance and potential use as a biological weapon. Proper identification, prevention strategies, and medical countermeasures are crucial in mitigating the risks associated with this bacterium.

Myeloid progenitor cells are a type of precursor cells that originate from hematopoietic stem cells (HSCs) in the bone marrow. These cells have the ability to differentiate into various types of blood cells, including red blood cells, platelets, and different kinds of white blood cells, specifically granulocytes (neutrophils, eosinophils, and basophils), monocytes, and megakaryocytes. Myeloid progenitor cells are crucial for the maintenance of normal hematopoiesis and immune function. Abnormalities in myeloid progenitor cell differentiation or function can lead to various hematological disorders such as leukemia, myelodysplastic syndromes, and myeloproliferative neoplasms.

Complement receptor 3b (CR3b or CD11b/CD18) is not a medical definition itself, but I can provide you with the relevant information regarding this term.

Complement receptor 3 (CR3) is a heterodimeric receptor consisting of two subunits, CD11b (also known as Mac-1 or CR3 alpha) and CD18 (also known as beta2 integrin). There are two forms of the CD11b/CD18 heterodimer: CR3a (CD11b/CD18) and CR3b (CD11b/CD18'). The difference between these two forms lies in the conformation of the CD11b subunit.

Complement receptor 3b (CR3b or CD11b/CD18') is a less common form of the CR3 receptor, which is primarily expressed on myeloid cells such as monocytes, macrophages, and neutrophils. CR3b has a higher affinity for complement component C3b and its fragments iC3b and C3dg compared to CR3a.

CR3b plays a role in various immune functions, including:

1. Phagocytosis: Binding of C3b or its fragments to CR3b facilitates the recognition and uptake of opsonized pathogens by phagocytes.
2. Adhesion: The integrin component of CR3b mediates cell-cell and cell-matrix interactions, contributing to leukocyte migration and recruitment to sites of inflammation or infection.
3. Intracellular signaling: Activation of CR3b can lead to intracellular signaling events that modulate immune responses, such as the release of pro-inflammatory cytokines and reactive oxygen species.

In summary, Complement receptor 3b (CR3b or CD11b/CD18') is a less common form of CR3 primarily expressed on myeloid cells that binds complement component C3b and its fragments with high affinity, mediating phagocytosis, adhesion, and intracellular signaling.

REceptor Activator of NF-kB (RANK) Ligand is a type of protein that plays a crucial role in the immune system and bone metabolism. It belongs to the tumor necrosis factor (TNF) superfamily and is primarily produced by osteoblasts, which are cells responsible for bone formation.

RANK Ligand binds to its receptor RANK, which is found on the surface of osteoclasts, a type of cell involved in bone resorption or breakdown. The binding of RANK Ligand to RANK activates signaling pathways that promote the differentiation, activation, and survival of osteoclasts, thereby increasing bone resorption.

Abnormalities in the RANKL-RANK signaling pathway have been implicated in various bone diseases, such as osteoporosis, rheumatoid arthritis, and certain types of cancer that metastasize to bones. Therefore, targeting this pathway with therapeutic agents has emerged as a promising approach for the treatment of these conditions.

Interferon Regulatory Factors (IRFs) are a family of transcription factors that play crucial roles in the regulation of immune responses, particularly in the expression of interferons (IFNs) and other genes involved in innate immunity and inflammation. In humans, there are nine known IRF proteins (IRF1-9), each with distinct functions and patterns of expression.

The primary function of IRFs is to regulate the transcription of type I IFNs (IFN-α and IFN-β) and other immune response genes in response to various stimuli, such as viral infections, bacterial components, and proinflammatory cytokines. IRFs can either activate or repress gene expression by binding to specific DNA sequences called interferon-stimulated response elements (ISREs) and/or IFN consensus sequences (ICSs) in the promoter regions of target genes.

IRF1, IRF3, and IRF7 are primarily involved in type I IFN regulation, with IRF1 acting as a transcriptional activator for IFN-β and various ISRE-containing genes, while IRF3 and IRF7 function as master regulators of the type I IFN response to viral infections. Upon viral recognition by pattern recognition receptors (PRRs), IRF3 and IRF7 are activated through phosphorylation and translocate to the nucleus, where they induce the expression of type I IFNs and other antiviral genes.

IRF2, IRF4, IRF5, and IRF8 have more diverse roles in immune regulation, including the control of T-cell differentiation, B-cell development, and myeloid cell function. For example, IRF4 is essential for the development and function of Th2 cells, while IRF5 and IRF8 are involved in the differentiation of dendritic cells and macrophages.

IRF6 and IRF9 have unique functions compared to other IRFs. IRF6 is primarily involved in epithelial cell development and differentiation, while IRF9 forms a complex with STAT1 and STAT2 to regulate the transcription of IFN-stimulated genes (ISGs) during the type I IFN response.

In summary, IRFs are a family of transcription factors that play crucial roles in various aspects of immune regulation, including antiviral responses, T-cell and B-cell development, and myeloid cell function. Dysregulation of IRF activity can lead to the development of autoimmune diseases, chronic inflammation, and cancer.

An acute disease is a medical condition that has a rapid onset, develops quickly, and tends to be short in duration. Acute diseases can range from minor illnesses such as a common cold or flu, to more severe conditions such as pneumonia, meningitis, or a heart attack. These types of diseases often have clear symptoms that are easy to identify, and they may require immediate medical attention or treatment.

Acute diseases are typically caused by an external agent or factor, such as a bacterial or viral infection, a toxin, or an injury. They can also be the result of a sudden worsening of an existing chronic condition. In general, acute diseases are distinct from chronic diseases, which are long-term medical conditions that develop slowly over time and may require ongoing management and treatment.

Examples of acute diseases include:

* Acute bronchitis: a sudden inflammation of the airways in the lungs, often caused by a viral infection.
* Appendicitis: an inflammation of the appendix that can cause severe pain and requires surgical removal.
* Gastroenteritis: an inflammation of the stomach and intestines, often caused by a viral or bacterial infection.
* Migraine headaches: intense headaches that can last for hours or days, and are often accompanied by nausea, vomiting, and sensitivity to light and sound.
* Myocardial infarction (heart attack): a sudden blockage of blood flow to the heart muscle, often caused by a buildup of plaque in the coronary arteries.
* Pneumonia: an infection of the lungs that can cause coughing, chest pain, and difficulty breathing.
* Sinusitis: an inflammation of the sinuses, often caused by a viral or bacterial infection.

It's important to note that while some acute diseases may resolve on their own with rest and supportive care, others may require medical intervention or treatment to prevent complications and promote recovery. If you are experiencing symptoms of an acute disease, it is always best to seek medical attention to ensure proper diagnosis and treatment.

Fluorescein-5-isothiocyanate (FITC) is not a medical term per se, but a chemical compound commonly used in biomedical research and clinical diagnostics. Therefore, I will provide a general definition of this term:

Fluorescein-5-isothiocyanate (FITC) is a fluorescent dye with an absorption maximum at approximately 492-495 nm and an emission maximum at around 518-525 nm. It is widely used as a labeling reagent for various biological molecules, such as antibodies, proteins, and nucleic acids, to study their structure, function, and interactions in techniques like flow cytometry, immunofluorescence microscopy, and western blotting. The isothiocyanate group (-N=C=S) in the FITC molecule reacts with primary amines (-NH2) present in biological molecules to form a stable thiourea bond, enabling specific labeling of target molecules for detection and analysis.

Paracrine communication is a form of cell-to-cell communication in which a cell releases a signaling molecule, known as a paracrine factor, that acts on nearby cells within the local microenvironment. This type of communication allows for the coordination and regulation of various cellular processes, including growth, differentiation, and survival.

Paracrine factors can be released from a cell through various mechanisms, such as exocytosis or diffusion through the extracellular matrix. Once released, these factors bind to specific receptors on the surface of nearby cells, triggering intracellular signaling pathways that lead to changes in gene expression and cell behavior.

Paracrine communication is an important mechanism for maintaining tissue homeostasis and coordinating responses to injury or disease. For example, during wound healing, paracrine signals released by immune cells can recruit other cells to the site of injury and stimulate their proliferation and differentiation to promote tissue repair.

It's worth noting that paracrine communication should be distinguished from autocrine signaling, where a cell releases a signaling molecule that binds back to its own receptors, and endocrine signaling, where a hormone is released into the bloodstream and travels to distant target cells.

Suppressors of Cytokine Signaling (SOCS) proteins are a family of intracellular signaling molecules that play a crucial role in regulating cytokine signaling pathways. They function as negative feedback inhibitors, helping to control the duration and intensity of cytokine responses.

There are eight known members of the SOCS family (SOCS1-7 and CIS), all of which share a similar structure consisting of:

1. An N-terminal domain, which varies among different SOCS proteins and is involved in specific target recognition.
2. A central SH2 (Src homology 2) domain, responsible for binding to phosphorylated tyrosine residues on cytokine receptors or other signaling molecules.
3. A C-terminal SOCS box, which serves as a protein-protein interaction module that recruits E3 ubiquitin ligases, leading to the degradation of target proteins via the ubiquitin-proteasome pathway.

SOCS proteins regulate cytokine signaling by inhibiting key components of the JAK-STAT (Janus kinase-signal transducer and activator of transcription) pathway, one of the major intracellular signaling cascades activated by cytokines. Specifically, SOCS1 and SOCS3 bind directly to the activated JAK kinases, preventing their interaction with STAT proteins and thus inhibiting downstream signal transduction. Additionally, SOCS proteins can also target receptors or JAKs for degradation via ubiquitination, further dampening cytokine signaling.

Dysregulation of SOCS protein expression has been implicated in various pathological conditions, including inflammatory diseases, autoimmune disorders, and cancer.

Tularemia is a bacterial disease caused by the gram-negative, facultatively intracellular bacterium Francisella tularensis. It is a zoonotic disease, meaning it primarily affects animals, but can also be transmitted to humans through various modes of exposure such as contact with infected animals or their tissues, ingestion of contaminated food or water, inhalation of infective aerosols, or bites from infected arthropods.

Humans typically develop symptoms within 3-5 days after exposure, which can vary depending on the route of infection and the specific Francisella tularensis subspecies involved. Common manifestations include fever, chills, headache, muscle aches, and fatigue. Depending on the type of tularemia, other symptoms may include skin ulcers, swollen lymph nodes, cough, chest pain, or diarrhea.

Tularemia is often classified into different clinical forms based on the route of infection and the initial site of multiplication:

1. Ulceroglandular tularemia: This form results from the bite of an infected arthropod (e.g., tick or deer fly) or contact with contaminated animal tissues, leading to a skin ulcer at the site of infection and swollen lymph nodes.
2. Glandular tularemia: Similar to ulceroglandular tularemia but without an obvious skin ulcer.
3. Oculoglandular tularemia: This form occurs when the bacteria come into contact with the eye, causing a painful inflammation of the eyelid and conjunctiva, along with swollen lymph nodes.
4. Oropharyngeal tularemia: Ingestion of contaminated food or water can lead to this form, characterized by sore throat, mouth ulcers, and swollen lymph nodes in the neck.
5. Pneumonic tularemia: This form results from inhalation of infective aerosols and is often associated with severe respiratory symptoms such as cough, chest pain, and pneumonia.
6. Typhoidal tularemia: A rare and severe form characterized by fever, rash, and systemic infection without any localizing signs or symptoms.

Tularemia is a serious bacterial infection that can be transmitted to humans through various routes, including insect bites, contact with contaminated animal tissues, ingestion of contaminated food or water, and inhalation of infective aerosols. Prompt diagnosis and appropriate antibiotic treatment are crucial for successful management of this potentially life-threatening disease.

Mitogen-Activated Protein Kinase 3 (MAPK3), also known as extracellular signal-regulated kinase 1 (ERK1), is a serine/threonine protein kinase that plays a crucial role in intracellular signal transduction pathways. It is involved in the regulation of various cellular processes, including proliferation, differentiation, and survival, in response to extracellular stimuli such as growth factors, hormones, and stress.

MAPK3 is activated through a phosphorylation cascade that involves the activation of upstream MAPK kinases (MKK or MEK). Once activated, MAPK3 can phosphorylate and activate various downstream targets, including transcription factors, to regulate gene expression. Dysregulation of MAPK3 signaling has been implicated in several diseases, including cancer and neurological disorders.

Complementary DNA (cDNA) is a type of DNA that is synthesized from a single-stranded RNA molecule through the process of reverse transcription. In this process, the enzyme reverse transcriptase uses an RNA molecule as a template to synthesize a complementary DNA strand. The resulting cDNA is therefore complementary to the original RNA molecule and is a copy of its coding sequence, but it does not contain non-coding regions such as introns that are present in genomic DNA.

Complementary DNA is often used in molecular biology research to study gene expression, protein function, and other genetic phenomena. For example, cDNA can be used to create cDNA libraries, which are collections of cloned cDNA fragments that represent the expressed genes in a particular cell type or tissue. These libraries can then be screened for specific genes or gene products of interest. Additionally, cDNA can be used to produce recombinant proteins in heterologous expression systems, allowing researchers to study the structure and function of proteins that may be difficult to express or purify from their native sources.

Phase-contrast microscopy is a type of optical microscopy that allows visualization of transparent or translucent specimens, such as living cells and their organelles, by increasing the contrast between areas with different refractive indices within the sample. This technique works by converting phase shifts in light passing through the sample into changes in amplitude, which can then be observed as differences in brightness and contrast.

In a phase-contrast microscope, a special condenser and objective are used to create an optical path difference between the direct and diffracted light rays coming from the specimen. The condenser introduces a phase shift for the diffracted light, while the objective contains a phase ring that compensates for this shift in the direct light. This results in the direct light appearing brighter than the diffracted light, creating contrast between areas with different refractive indices within the sample.

Phase-contrast microscopy is particularly useful for observing unstained living cells and their dynamic processes, such as cell division, motility, and secretion, without the need for stains or dyes that might affect their viability or behavior.

"SRS-A" is an older abbreviation for "Slow-Reacting Substance of Anaphylaxis," which refers to a group of molecules called "leukotrienes." Leukotrienes are mediators of inflammation and play a key role in the pathogenesis of asthma and other allergic diseases. They are produced by mast cells and basophils upon activation, and cause bronchoconstriction, increased vascular permeability, and mucus production.

The term "SRS-A" is not commonly used in modern medical literature, as it has been largely replaced by the more specific names of its individual components: LTC4, LTD4, and LTE4. These leukotrienes are now collectively referred to as the "cysteinyl leukotrienes."

Blood bactericidal activity refers to the ability of an individual's blood to kill or inhibit the growth of bacteria. This is an important aspect of the body's immune system, as it helps to prevent infection and maintain overall health. The bactericidal activity of blood can be influenced by various factors, including the presence of antibodies, white blood cells (such as neutrophils), and complement proteins.

In medical terms, the term "bactericidal" specifically refers to an agent or substance that is capable of killing bacteria. Therefore, when we talk about blood bactericidal activity, we are referring to the collective ability of various components in the blood to kill or inhibit the growth of bacteria. This is often measured in laboratory tests as a way to assess a person's immune function and their susceptibility to infection.

It's worth noting that not all substances in the blood are bactericidal; some may simply inhibit the growth of bacteria without killing them. These substances are referred to as bacteriostatic. Both bactericidal and bacteriostatic agents play important roles in maintaining the body's defense against infection.

Thymidine is a pyrimidine nucleoside that consists of a thymine base linked to a deoxyribose sugar by a β-N1-glycosidic bond. It plays a crucial role in DNA replication and repair processes as one of the four nucleosides in DNA, along with adenosine, guanosine, and cytidine. Thymidine is also used in research and clinical settings for various purposes, such as studying DNA synthesis or as a component of antiviral and anticancer therapies.

Complement C5a is a protein fragment that is generated during the activation of the complement system, which is a part of the immune system. The complement system helps to eliminate pathogens and damaged cells from the body by tagging them for destruction and attracting immune cells to the site of infection or injury.

C5a is formed when the fifth component of the complement system (C5) is cleaved into two smaller fragments, C5a and C5b, during the complement activation cascade. C5a is a potent pro-inflammatory mediator that can attract and activate various immune cells, such as neutrophils, monocytes, and eosinophils, to the site of infection or injury. It can also increase vascular permeability, promote the release of histamine, and induce the production of reactive oxygen species, all of which contribute to the inflammatory response.

However, excessive or uncontrolled activation of the complement system and generation of C5a can lead to tissue damage and inflammation, contributing to the pathogenesis of various diseases, such as sepsis, acute respiratory distress syndrome (ARDS), and autoimmune disorders. Therefore, targeting C5a or its receptors has been explored as a potential therapeutic strategy for these conditions.

CD8-positive T-lymphocytes, also known as CD8+ T cells or cytotoxic T cells, are a type of white blood cell that plays a crucial role in the adaptive immune system. They are named after the CD8 molecule found on their surface, which is a protein involved in cell signaling and recognition.

CD8+ T cells are primarily responsible for identifying and destroying virus-infected cells or cancerous cells. When activated, they release cytotoxic granules that contain enzymes capable of inducing apoptosis (programmed cell death) in the target cells. They also produce cytokines such as interferon-gamma, which can help coordinate the immune response and activate other immune cells.

CD8+ T cells are generated in the thymus gland and are a type of T cell, which is a lymphocyte that matures in the thymus and plays a central role in cell-mediated immunity. They recognize and respond to specific antigens presented on the surface of infected or cancerous cells in conjunction with major histocompatibility complex (MHC) class I molecules.

Overall, CD8+ T cells are an essential component of the immune system's defense against viral infections and cancer.

Glucans are polysaccharides (complex carbohydrates) that are made up of long chains of glucose molecules. They can be found in the cell walls of certain plants, fungi, and bacteria. In medicine, beta-glucans derived from yeast or mushrooms have been studied for their potential immune-enhancing effects. However, more research is needed to fully understand their role and effectiveness in human health.

Toxoplasmosis is a zoonotic disease, meaning it can be transmitted from animals to humans. It is caused by the intracellular protozoan parasite Toxoplasma gondii. This parasite can infect a wide range of warm-blooded animals, including birds and mammals, as intermediate hosts. However, cats are the primary definitive host for this parasite because the sexual stage of the parasite's life cycle occurs in their intestines, leading to the shedding of oocysts (environmentally resistant stages) in their feces.

Animals can become infected with Toxoplasma gondii through several routes:

1. Ingestion of sporulated oocysts from contaminated soil, water, or food.
2. Consumption of tissue cysts present in the tissues of infected animals during predation.
3. Vertical transmission (transplacental) from an infected mother to her offspring.

Clinical signs and symptoms of toxoplasmosis in animals can vary depending on their age, immune status, and the parasite's virulence. In many cases, animals may not show any apparent signs of infection, but some may develop:

1. Generalized illness with fever, lethargy, and loss of appetite.
2. Lymphadenopathy (swollen lymph nodes).
3. Neurological symptoms such as tremors, ataxia (lack of coordination), or seizures if the central nervous system is affected.
4. Eye lesions, including inflammation and scarring of the retina, which can lead to vision loss in severe cases.
5. Reproductive issues, such as abortion, stillbirths, or birth defects in offspring when pregnant females are infected.

It is important to note that while toxoplasmosis can cause significant health problems in animals, particularly in immunocompromised individuals and developing fetuses, it is often asymptomatic or mild in healthy adult animals. Nonetheless, the zoonotic potential of Toxoplasma gondii highlights the importance of practicing good hygiene and taking necessary precautions when handling infected animals or their waste to minimize the risk of transmission to humans.

Sarcoidosis is a multi-system disorder characterized by the formation of granulomas (small clumps of inflammatory cells) in various organs, most commonly the lungs and lymphatic system. These granulomas can impair the function of the affected organ(s), leading to a variety of symptoms. The exact cause of sarcoidosis is unknown, but it's thought to be an overactive immune response to an unknown antigen, possibly triggered by an infection, chemical exposure, or another environmental factor.

The diagnosis of sarcoidosis typically involves a combination of clinical evaluation, imaging studies (such as chest X-rays and CT scans), and laboratory tests (including blood tests and biopsies). While there is no cure for sarcoidosis, treatment may be necessary to manage symptoms and prevent complications. Corticosteroids are often used to suppress the immune system and reduce inflammation, while other medications may be prescribed to treat specific organ involvement or symptoms. In some cases, sarcoidosis may resolve on its own without any treatment.

Metalloendopeptidases are a type of enzymes that cleave peptide bonds in proteins, specifically at interior positions within the polypeptide chain. They require metal ions as cofactors for their catalytic activity, typically zinc (Zn2+) or cobalt (Co2+). These enzymes play important roles in various biological processes such as protein degradation, processing, and signaling. Examples of metalloendopeptidases include thermolysin, matrix metalloproteinases (MMPs), and neutrophil elastase.

Molecular cloning is a laboratory technique used to create multiple copies of a specific DNA sequence. This process involves several steps:

1. Isolation: The first step in molecular cloning is to isolate the DNA sequence of interest from the rest of the genomic DNA. This can be done using various methods such as PCR (polymerase chain reaction), restriction enzymes, or hybridization.
2. Vector construction: Once the DNA sequence of interest has been isolated, it must be inserted into a vector, which is a small circular DNA molecule that can replicate independently in a host cell. Common vectors used in molecular cloning include plasmids and phages.
3. Transformation: The constructed vector is then introduced into a host cell, usually a bacterial or yeast cell, through a process called transformation. This can be done using various methods such as electroporation or chemical transformation.
4. Selection: After transformation, the host cells are grown in selective media that allow only those cells containing the vector to grow. This ensures that the DNA sequence of interest has been successfully cloned into the vector.
5. Amplification: Once the host cells have been selected, they can be grown in large quantities to amplify the number of copies of the cloned DNA sequence.

Molecular cloning is a powerful tool in molecular biology and has numerous applications, including the production of recombinant proteins, gene therapy, functional analysis of genes, and genetic engineering.

Pulmonary Alveolar Proteinosis (PAP) is a rare lung disorder characterized by the accumulation of surfactant, a lipoprotein complex that reduces surface tension within the alveoli, in the air sacs (alveoli) of the lungs. This accumulation can lead to difficulty breathing and reduced oxygen levels in the blood.

There are three types of PAP:

1. Congenital PAP: A very rare inherited form that affects infants and is caused by a genetic mutation that disrupts the production or function of granulocyte-macrophage colony-stimulating factor (GM-CSF), a protein important for the development and function of alveolar macrophages.

2. Secondary PAP: This form is associated with conditions that impair the clearance of surfactant by alveolar macrophages, such as hematologic disorders (e.g., leukemia), infections, exposure to inhaled irritants (e.g., silica dust), and certain medications.

3. Idiopathic PAP: The most common form, also known as autoimmune PAP, is caused by the development of autoantibodies against GM-CSF, which disrupts its function and leads to surfactant accumulation in the lungs.

Treatment for PAP may include whole lung lavage (WLL), a procedure where the affected lung is filled with saline solution and then drained to remove excess surfactant, as well as managing any underlying conditions. In some cases of idiopathic PAP, off-label use of inhaled GM-CSF has shown promise in improving symptoms and lung function.

Intracellular signaling peptides and proteins are molecules that play a crucial role in transmitting signals within cells, which ultimately lead to changes in cell behavior or function. These signals can originate from outside the cell (extracellular) or within the cell itself. Intracellular signaling molecules include various types of peptides and proteins, such as:

1. G-protein coupled receptors (GPCRs): These are seven-transmembrane domain receptors that bind to extracellular signaling molecules like hormones, neurotransmitters, or chemokines. Upon activation, they initiate a cascade of intracellular signals through G proteins and secondary messengers.
2. Receptor tyrosine kinases (RTKs): These are transmembrane receptors that bind to growth factors, cytokines, or hormones. Activation of RTKs leads to autophosphorylation of specific tyrosine residues, creating binding sites for intracellular signaling proteins such as adapter proteins, phosphatases, and enzymes like Ras, PI3K, and Src family kinases.
3. Second messenger systems: Intracellular second messengers are small molecules that amplify and propagate signals within the cell. Examples include cyclic adenosine monophosphate (cAMP), cyclic guanosine monophosphate (cGMP), diacylglycerol (DAG), inositol triphosphate (IP3), calcium ions (Ca2+), and nitric oxide (NO). These second messengers activate or inhibit various downstream effectors, leading to changes in cellular responses.
4. Signal transduction cascades: Intracellular signaling proteins often form complex networks of interacting molecules that relay signals from the plasma membrane to the nucleus. These cascades involve kinases (protein kinases A, B, C, etc.), phosphatases, and adapter proteins, which ultimately regulate gene expression, cell cycle progression, metabolism, and other cellular processes.
5. Ubiquitination and proteasome degradation: Intracellular signaling pathways can also control protein stability by modulating ubiquitin-proteasome degradation. E3 ubiquitin ligases recognize specific substrates and conjugate them with ubiquitin molecules, targeting them for proteasomal degradation. This process regulates the abundance of key signaling proteins and contributes to signal termination or amplification.

In summary, intracellular signaling pathways involve a complex network of interacting proteins that relay signals from the plasma membrane to various cellular compartments, ultimately regulating gene expression, metabolism, and other cellular processes. Dysregulation of these pathways can contribute to disease development and progression, making them attractive targets for therapeutic intervention.

Interleukin-2 (IL-2) is a type of cytokine, which are signaling molecules that mediate and regulate immunity, inflammation, and hematopoiesis. Specifically, IL-2 is a growth factor for T cells, a type of white blood cell that plays a central role in the immune response. It is primarily produced by CD4+ T cells (also known as T helper cells) and stimulates the proliferation and differentiation of activated T cells, including effector T cells and regulatory T cells. IL-2 also has roles in the activation and function of other immune cells, such as B cells, natural killer cells, and dendritic cells. Dysregulation of IL-2 production or signaling can contribute to various pathological conditions, including autoimmune diseases, chronic infections, and cancer.

Immunosuppression is a state in which the immune system's ability to mount an immune response is reduced, compromised or inhibited. This can be caused by certain medications (such as those used to prevent rejection of transplanted organs), diseases (like HIV/AIDS), or genetic disorders. As a result, the body becomes more susceptible to infections and cancer development. It's important to note that immunosuppression should not be confused with immunity, which refers to the body's ability to resist and fight off infections and diseases.

Peptides are short chains of amino acid residues linked by covalent bonds, known as peptide bonds. They are formed when two or more amino acids are joined together through a condensation reaction, which results in the elimination of a water molecule and the formation of an amide bond between the carboxyl group of one amino acid and the amino group of another.

Peptides can vary in length from two to about fifty amino acids, and they are often classified based on their size. For example, dipeptides contain two amino acids, tripeptides contain three, and so on. Oligopeptides typically contain up to ten amino acids, while polypeptides can contain dozens or even hundreds of amino acids.

Peptides play many important roles in the body, including serving as hormones, neurotransmitters, enzymes, and antibiotics. They are also used in medical research and therapeutic applications, such as drug delivery and tissue engineering.

"Blood physiological phenomena" is a broad term that refers to various functions, processes, and characteristics related to the blood in the body. Here are some definitions of specific blood-related physiological phenomena:

1. Hematopoiesis: The process of producing blood cells in the bone marrow. This includes the production of red blood cells (erythropoiesis), white blood cells (leukopoiesis), and platelets (thrombopoiesis).
2. Hemostasis: The body's response to stop bleeding or prevent excessive blood loss after injury. It involves a complex interplay between blood vessels, platelets, and clotting factors that work together to form a clot.
3. Osmoregulation: The regulation of water and electrolyte balance in the blood. This is achieved through various mechanisms such as thirst, urine concentration, and hormonal control.
4. Acid-base balance: The maintenance of a stable pH level in the blood. This involves the balance between acidic and basic components in the blood, which can be affected by factors such as respiration, metabolism, and kidney function.
5. Hemoglobin function: The ability of hemoglobin molecules in red blood cells to bind and transport oxygen from the lungs to tissues throughout the body.
6. Blood viscosity: The thickness or flowability of blood, which can affect its ability to circulate through the body. Factors that can influence blood viscosity include hematocrit (the percentage of red blood cells in the blood), plasma proteins, and temperature.
7. Immunological function: The role of white blood cells and other components of the immune system in protecting the body against infection and disease. This includes the production of antibodies, phagocytosis (the engulfing and destruction of foreign particles), and inflammation.

RNA interference (RNAi) is a biological process in which RNA molecules inhibit the expression of specific genes. This process is mediated by small RNA molecules, including microRNAs (miRNAs) and small interfering RNAs (siRNAs), that bind to complementary sequences on messenger RNA (mRNA) molecules, leading to their degradation or translation inhibition.

RNAi plays a crucial role in regulating gene expression and defending against foreign genetic elements, such as viruses and transposons. It has also emerged as an important tool for studying gene function and developing therapeutic strategies for various diseases, including cancer and viral infections.

CD11 is a group of integrin proteins that are present on the surface of various immune cells, including neutrophils, monocytes, and macrophages. They play a crucial role in the adhesion and migration of these cells to sites of inflammation or injury. CD11 includes three distinct subunits: CD11a (also known as LFA-1), CD11b (also known as Mac-1 or Mo1), and CD11c (also known as p150,95).

Antigens are substances that can stimulate an immune response in the body. In the context of CD11, antigens may refer to specific molecules or structures on pathogens such as bacteria or viruses that can be recognized by CD11-expressing immune cells. These antigens bind to CD11 and trigger a series of intracellular signaling events that lead to the activation and migration of the immune cells to the site of infection or injury.

Therefore, the medical definition of 'antigens, CD11' may refer to specific molecules or structures on pathogens that can bind to CD11 proteins on immune cells and trigger an immune response.

Interferon receptors are cell surface proteins that bind to interferons, which are a group of signaling proteins made and released by host cells in response to the presence of viruses, parasites, or tumor cells. These receptors belong to the class II cytokine receptor family and are found on the membranes of many cell types, including leukocytes, fibroblasts, and endothelial cells.

There are two main types of interferon receptors: type I and type II. Type I interferon receptors (IFNAR) bind to type I interferons (IFN-α, IFN-β, and IFN-ω), while type II interferon receptors (IFNGR) bind to type II interferon (IFN-γ).

Once interferons bind to their respective receptors, they activate a signaling cascade that leads to the expression of genes involved in the immune response, such as those encoding antiviral proteins and cytokines. This helps to protect cells from viral infection and modulate the immune system's response to threats.

Interferon receptors play an essential role in the body's defense against infectious diseases and cancer. Dysregulation of interferon signaling has been implicated in various pathological conditions, including autoimmune disorders and viral infections that evade the immune system.

The intracellular space refers to the interior of a cell, specifically the area enclosed by the plasma membrane that is occupied by organelles, cytoplasm, and other cellular structures. It excludes the extracellular space, which is the area outside the cell surrounded by the plasma membrane. The intracellular space is where various metabolic processes, such as protein synthesis, energy production, and waste removal, occur. It is essential for maintaining the cell's structure, function, and survival.

Antibody formation, also known as humoral immune response, is the process by which the immune system produces proteins called antibodies in response to the presence of a foreign substance (antigen) in the body. This process involves several steps:

1. Recognition: The antigen is recognized and bound by a type of white blood cell called a B lymphocyte or B cell, which then becomes activated.
2. Differentiation: The activated B cell undergoes differentiation to become a plasma cell, which is a type of cell that produces and secretes large amounts of antibodies.
3. Antibody production: The plasma cells produce and release antibodies, which are proteins made up of four polypeptide chains (two heavy chains and two light chains) arranged in a Y-shape. Each antibody has two binding sites that can recognize and bind to specific regions on the antigen called epitopes.
4. Neutralization or elimination: The antibodies bind to the antigens, neutralizing them or marking them for destruction by other immune cells. This helps to prevent the spread of infection and protect the body from harmful substances.

Antibody formation is an important part of the adaptive immune response, which allows the body to specifically recognize and respond to a wide variety of pathogens and foreign substances.

Chemokine (C-X-C motif) ligand 10 (CXCL10), also known as interferon-gamma-inducible protein 10 (IP-10), is a small cytokine protein that belongs to the chemokine family. Chemokines are a group of signaling proteins that play crucial roles in immune responses and inflammation by recruiting various immune cells to the sites of infection or injury.

CXCL10 is primarily produced by several cell types, including monocytes, endothelial cells, and fibroblasts, in response to stimulation by interferon-gamma (IFN-γ), a cytokine that is critical for the activation of immune cells during an immune response. CXCL10 specifically binds to and activates its receptor, CXCR3, which is expressed on various immune cells such as T lymphocytes, natural killer (NK) cells, and monocytes.

The binding of CXCL10 to CXCR3 triggers a cascade of intracellular signaling events that result in the activation and migration of these immune cells towards the site of inflammation or infection. Consequently, CXCL10 plays essential roles in various physiological and pathological processes, including the recruitment of immune cells to sites of viral infections, tumor growth, and autoimmune diseases.

In summary, Chemokine CXCL10 is a crucial signaling protein that mediates immune cell trafficking and activation during inflammation and immune responses.

NF-κB (Nuclear Factor kappa-light-chain-enhancer of activated B cells) p50 subunit, also known as NFKB1, is a protein that plays a crucial role in regulating the immune response, inflammation, and cell survival. The NF-κB p50 subunit can form homodimers or heterodimers with other NF-κB family members, such as p65 (RelA) or c-Rel, to bind to specific DNA sequences called κB sites in the promoter regions of target genes.

The activation of NF-κB signaling leads to the nuclear translocation of these dimers and the regulation of gene expression involved in various biological processes, including immune response, inflammation, differentiation, cell growth, and apoptosis. The p50 subunit can act as a transcriptional activator or repressor, depending on its partner and the context.

In summary, NF-κB p50 Subunit is a protein involved in the regulation of gene expression, particularly in immune response, inflammation, and cell survival, through its ability to bind to specific DNA sequences as part of homodimers or heterodimers with other NF-κB family members.

The intestinal mucosa is the innermost layer of the intestines, which comes into direct contact with digested food and microbes. It is a specialized epithelial tissue that plays crucial roles in nutrient absorption, barrier function, and immune defense. The intestinal mucosa is composed of several cell types, including absorptive enterocytes, mucus-secreting goblet cells, hormone-producing enteroendocrine cells, and immune cells such as lymphocytes and macrophages.

The surface of the intestinal mucosa is covered by a single layer of epithelial cells, which are joined together by tight junctions to form a protective barrier against harmful substances and microorganisms. This barrier also allows for the selective absorption of nutrients into the bloodstream. The intestinal mucosa also contains numerous lymphoid follicles, known as Peyer's patches, which are involved in immune surveillance and defense against pathogens.

In addition to its role in absorption and immunity, the intestinal mucosa is also capable of producing hormones that regulate digestion and metabolism. Dysfunction of the intestinal mucosa can lead to various gastrointestinal disorders, such as inflammatory bowel disease, celiac disease, and food allergies.

'Cell lineage' is a term used in biology and medicine to describe the developmental history or relationship of a cell or group of cells to other cells, tracing back to the original progenitor or stem cell. It refers to the series of cell divisions and differentiation events that give rise to specific types of cells in an organism over time.

In simpler terms, cell lineage is like a family tree for cells, showing how they are related to each other through a chain of cell division and specialization events. This concept is important in understanding the development, growth, and maintenance of tissues and organs in living beings.

Cytotoxicity tests, immunologic are a group of laboratory assays used to measure the immune-mediated damage or destruction (cytotoxicity) of cells. These tests are often used in medical research and clinical settings to evaluate the potential toxicity of drugs, biological agents, or environmental factors on specific types of cells.

Immunologic cytotoxicity tests typically involve the use of immune effector cells, such as cytotoxic T lymphocytes (CTLs) or natural killer (NK) cells, which can recognize and kill target cells that express specific antigens on their surface. The tests may also involve the use of antibodies or other immune molecules that can bind to target cells and trigger complement-mediated cytotoxicity.

There are several types of immunologic cytotoxicity tests, including:

1. Cytotoxic T lymphocyte (CTL) assays: These tests measure the ability of CTLs to recognize and kill target cells that express specific antigens. The test involves incubating target cells with CTLs and then measuring the amount of cell death or damage.
2. Natural killer (NK) cell assays: These tests measure the ability of NK cells to recognize and kill target cells that lack self-antigens or express stress-induced antigens. The test involves incubating target cells with NK cells and then measuring the amount of cell death or damage.
3. Antibody-dependent cellular cytotoxicity (ADCC) assays: These tests measure the ability of antibodies to bind to target cells and recruit immune effector cells, such as NK cells or macrophages, to mediate cell lysis. The test involves incubating target cells with antibodies and then measuring the amount of cell death or damage.
4. Complement-dependent cytotoxicity (CDC) assays: These tests measure the ability of complement proteins to bind to target cells and form a membrane attack complex that leads to cell lysis. The test involves incubating target cells with complement proteins and then measuring the amount of cell death or damage.

Immunologic cytotoxicity tests are important tools in immunology, cancer research, and drug development. They can help researchers understand how immune cells recognize and kill infected or damaged cells, as well as how to develop new therapies that enhance or inhibit these processes.

Imidazoles are a class of heterocyclic organic compounds that contain a double-bonded nitrogen atom and two additional nitrogen atoms in the ring. They have the chemical formula C3H4N2. In a medical context, imidazoles are commonly used as antifungal agents. Some examples of imidazole-derived antifungals include clotrimazole, miconazole, and ketoconazole. These medications work by inhibiting the synthesis of ergosterol, a key component of fungal cell membranes, leading to increased permeability and death of the fungal cells. Imidazoles may also have anti-inflammatory, antibacterial, and anticancer properties.

CD80 (also known as B7-1) is a cell surface protein that functions as a costimulatory molecule in the immune system. It is primarily expressed on antigen presenting cells such as dendritic cells, macrophages, and B cells. CD80 binds to the CD28 receptor on T cells, providing a critical second signal necessary for T cell activation and proliferation. This interaction plays a crucial role in the initiation of an effective immune response against pathogens and tumors.

CD80 can also interact with another receptor called CTLA-4 (cytotoxic T lymphocyte antigen 4), which is expressed on activated T cells. The binding of CD80 to CTLA-4 delivers a negative signal that helps regulate the immune response and prevent overactivation, contributing to the maintenance of self-tolerance and preventing autoimmunity.

In summary, CD80 is an important antigen involved in the regulation of the adaptive immune response by modulating T cell activation and proliferation through its interactions with CD28 and CTLA-4 receptors.

Antimicrobial cationic peptides (ACPs) are a group of small, naturally occurring peptides that possess broad-spectrum antimicrobial activity against various microorganisms, including bacteria, fungi, viruses, and parasites. They are called "cationic" because they contain positively charged amino acid residues (such as lysine and arginine), which allow them to interact with and disrupt the negatively charged membranes of microbial cells.

ACPs are produced by a wide range of organisms, including humans, animals, and plants, as part of their innate immune response to infection. They play an important role in protecting the host from invading pathogens by directly killing them or inhibiting their growth.

The antimicrobial activity of ACPs is thought to be mediated by their ability to disrupt the membranes of microbial cells, leading to leakage of cellular contents and death. Some ACPs may also have intracellular targets, such as DNA or protein synthesis, that contribute to their antimicrobial activity.

ACPs are being studied for their potential use as therapeutic agents to treat infectious diseases, particularly those caused by drug-resistant bacteria. However, their clinical application is still in the early stages of development due to concerns about their potential toxicity to host cells and the emergence of resistance mechanisms in microbial pathogens.

Thromboplastin is a substance that activates the coagulation cascade, leading to the formation of a clot (thrombus). It's primarily found in damaged or injured tissues and blood vessels, as well as in platelets (thrombocytes). There are two types of thromboplastin:

1. Extrinsic thromboplastin (also known as tissue factor): This is a transmembrane glycoprotein that is primarily found in subendothelial cells and released upon injury to the blood vessels. It initiates the extrinsic pathway of coagulation by binding to and activating Factor VII, ultimately leading to the formation of thrombin and fibrin clots.
2. Intrinsic thromboplastin (also known as plasma thromboplastin or factor III): This term is used less frequently and refers to a labile phospholipid component present in platelet membranes, which plays a role in the intrinsic pathway of coagulation.

In clinical settings, the term "thromboplastin" often refers to reagents used in laboratory tests like the prothrombin time (PT) and activated partial thromboplastin time (aPTT). These reagents contain a source of tissue factor and calcium ions to initiate and monitor the coagulation process.

A drug carrier, also known as a drug delivery system or vector, is a vehicle that transports a pharmaceutical compound to a specific site in the body. The main purpose of using drug carriers is to improve the efficacy and safety of drugs by enhancing their solubility, stability, bioavailability, and targeted delivery, while minimizing unwanted side effects.

Drug carriers can be made up of various materials, including natural or synthetic polymers, lipids, inorganic nanoparticles, or even cells and viruses. They can encapsulate, adsorb, or conjugate drugs through different mechanisms, such as physical entrapment, electrostatic interaction, or covalent bonding.

Some common types of drug carriers include:

1. Liposomes: spherical vesicles composed of one or more lipid bilayers that can encapsulate hydrophilic and hydrophobic drugs.
2. Polymeric nanoparticles: tiny particles made of biodegradable polymers that can protect drugs from degradation and enhance their accumulation in target tissues.
3. Dendrimers: highly branched macromolecules with a well-defined structure and size that can carry multiple drug molecules and facilitate their release.
4. Micelles: self-assembled structures formed by amphiphilic block copolymers that can solubilize hydrophobic drugs in water.
5. Inorganic nanoparticles: such as gold, silver, or iron oxide nanoparticles, that can be functionalized with drugs and targeting ligands for diagnostic and therapeutic applications.
6. Cell-based carriers: living cells, such as red blood cells, stem cells, or immune cells, that can be loaded with drugs and used to deliver them to specific sites in the body.
7. Viral vectors: modified viruses that can infect cells and introduce genetic material encoding therapeutic proteins or RNA interference molecules.

The choice of drug carrier depends on various factors, such as the physicochemical properties of the drug, the route of administration, the target site, and the desired pharmacokinetics and biodistribution. Therefore, selecting an appropriate drug carrier is crucial for achieving optimal therapeutic outcomes and minimizing side effects.

Septic shock is a serious condition that occurs as a complication of an infection that has spread throughout the body. It's characterized by a severe drop in blood pressure and abnormalities in cellular metabolism, which can lead to organ failure and death if not promptly treated.

In septic shock, the immune system overreacts to an infection, releasing an overwhelming amount of inflammatory chemicals into the bloodstream. This leads to widespread inflammation, blood vessel dilation, and leaky blood vessels, which can cause fluid to leak out of the blood vessels and into surrounding tissues. As a result, the heart may not be able to pump enough blood to vital organs, leading to organ failure.

Septic shock is often caused by bacterial infections, but it can also be caused by fungal or viral infections. It's most commonly seen in people with weakened immune systems, such as those who have recently undergone surgery, have chronic medical conditions, or are taking medications that suppress the immune system.

Prompt diagnosis and treatment of septic shock is critical to prevent long-term complications and improve outcomes. Treatment typically involves aggressive antibiotic therapy, intravenous fluids, vasopressors to maintain blood pressure, and supportive care in an intensive care unit (ICU).

Cryptococcosis is a fungal infection caused by the yeast-like fungus Cryptococcus neoformans or Cryptococcus gattii. It can affect people with weakened immune systems, such as those with HIV/AIDS, cancer, organ transplants, or long-term steroid use. The infection typically starts in the lungs and can spread to other parts of the body, including the brain (meningitis), causing various symptoms like cough, fever, chest pain, headache, confusion, and vision problems. Treatment usually involves antifungal medications, and the prognosis depends on the patient's immune status and the severity of the infection.

Antibody-Dependent Cell Cytotoxicity (ADCC) is a type of immune response in which the effector cells of the immune system, such as natural killer (NK) cells, cytotoxic T-cells or macrophages, recognize and destroy virus-infected or cancer cells that are coated with antibodies.

In this process, an antibody produced by B-cells binds specifically to an antigen on the surface of a target cell. The other end of the antibody then interacts with Fc receptors found on the surface of effector cells. This interaction triggers the effector cells to release cytotoxic substances, such as perforins and granzymes, which create pores in the target cell membrane and induce apoptosis (programmed cell death).

ADCC plays an important role in the immune defense against viral infections and cancer. It is also a mechanism of action for some monoclonal antibody therapies used in cancer treatment.

Toll-like receptor 3 (TLR3) is a type of protein belonging to the family of Toll-like receptors, which are involved in the innate immune system's response to pathogens. TLR3 is primarily expressed on the surface of various cells including immune cells such as dendritic cells, macrophages, and epithelial cells.

TLR3 recognizes double-stranded RNA (dsRNA), a molecule found in certain viruses during their replication process. When TLR3 binds to dsRNA, it triggers a signaling cascade that leads to the activation of several transcription factors, including NF-κB and IRF3, which ultimately result in the production of proinflammatory cytokines and type I interferons (IFNs). These molecules play crucial roles in activating the immune response against viral infections.

In summary, TLR3 is a pattern recognition receptor that plays an essential role in the early detection and defense against viral pathogens by initiating innate immune responses upon recognizing double-stranded RNA.

Electrophoresis, polyacrylamide gel (EPG) is a laboratory technique used to separate and analyze complex mixtures of proteins or nucleic acids (DNA or RNA) based on their size and electrical charge. This technique utilizes a matrix made of cross-linked polyacrylamide, a type of gel, which provides a stable and uniform environment for the separation of molecules.

In this process:

1. The polyacrylamide gel is prepared by mixing acrylamide monomers with a cross-linking agent (bis-acrylamide) and a catalyst (ammonium persulfate) in the presence of a buffer solution.
2. The gel is then poured into a mold and allowed to polymerize, forming a solid matrix with uniform pore sizes that depend on the concentration of acrylamide used. Higher concentrations result in smaller pores, providing better resolution for separating smaller molecules.
3. Once the gel has set, it is placed in an electrophoresis apparatus containing a buffer solution. Samples containing the mixture of proteins or nucleic acids are loaded into wells on the top of the gel.
4. An electric field is applied across the gel, causing the negatively charged molecules to migrate towards the positive electrode (anode) while positively charged molecules move toward the negative electrode (cathode). The rate of migration depends on the size, charge, and shape of the molecules.
5. Smaller molecules move faster through the gel matrix and will migrate farther from the origin compared to larger molecules, resulting in separation based on size. Proteins and nucleic acids can be selectively stained after electrophoresis to visualize the separated bands.

EPG is widely used in various research fields, including molecular biology, genetics, proteomics, and forensic science, for applications such as protein characterization, DNA fragment analysis, cloning, mutation detection, and quality control of nucleic acid or protein samples.

Mitogen-Activated Protein Kinase 1 (MAPK1), also known as Extracellular Signal-Regulated Kinase 2 (ERK2), is a protein kinase that plays a crucial role in intracellular signal transduction pathways. It is a member of the MAPK family, which regulates various cellular processes such as proliferation, differentiation, apoptosis, and stress response.

MAPK1 is activated by a cascade of phosphorylation events initiated by upstream activators like MAPKK (Mitogen-Activated Protein Kinase Kinase) in response to various extracellular signals such as growth factors, hormones, and mitogens. Once activated, MAPK1 phosphorylates downstream targets, including transcription factors and other protein kinases, thereby modulating their activities and ultimately influencing gene expression and cellular responses.

MAPK1 is widely expressed in various tissues and cells, and its dysregulation has been implicated in several pathological conditions, including cancer, inflammation, and neurodegenerative diseases. Therefore, understanding the regulation and function of MAPK1 signaling pathways has important implications for developing therapeutic strategies to treat these disorders.

Blocking antibodies are a type of antibody that binds to a specific antigen but does not cause the immune system to directly attack the antigen. Instead, blocking antibodies prevent the antigen from interacting with other molecules or receptors, effectively "blocking" its activity. This can be useful in therapeutic settings, where blocking antibodies can be used to inhibit the activity of harmful proteins or toxins.

For example, some blocking antibodies have been developed to target and block the activity of specific cytokines, which are signaling molecules involved in inflammation and immune responses. By blocking the interaction between the cytokine and its receptor, these antibodies can help to reduce inflammation and alleviate symptoms in certain autoimmune diseases or chronic inflammatory conditions.

It's important to note that while blocking antibodies can be useful for therapeutic purposes, they can also have unintended consequences if they block the activity of essential proteins or molecules. Therefore, careful consideration and testing are required before using blocking antibodies as a treatment.

A peptide fragment is a short chain of amino acids that is derived from a larger peptide or protein through various biological or chemical processes. These fragments can result from the natural breakdown of proteins in the body during regular physiological processes, such as digestion, or they can be produced experimentally in a laboratory setting for research or therapeutic purposes.

Peptide fragments are often used in research to map the structure and function of larger peptides and proteins, as well as to study their interactions with other molecules. In some cases, peptide fragments may also have biological activity of their own and can be developed into drugs or diagnostic tools. For example, certain peptide fragments derived from hormones or neurotransmitters may bind to receptors in the body and mimic or block the effects of the full-length molecule.

HL-60 cells are a type of human promyelocytic leukemia cell line that is commonly used in scientific research. They are named after the hospital where they were first isolated, the Hospital of the University of Pennsylvania (HUP) and the 60th culture attempt to grow these cells.

HL-60 cells have the ability to differentiate into various types of blood cells, such as granulocytes, monocytes, and macrophages, when exposed to certain chemical compounds or under specific culturing conditions. This makes them a valuable tool for studying the mechanisms of cell differentiation, proliferation, and apoptosis (programmed cell death).

HL-60 cells are also often used in toxicity studies, drug discovery and development, and research on cancer, inflammation, and infectious diseases. They can be easily grown in the lab and have a stable genotype, making them ideal for use in standardized experiments and comparisons between different studies.

Immunological models are simplified representations or simulations of the immune system's structure, function, and interactions with pathogens or other entities. These models can be theoretical (conceptual), mathematical, or computational and are used to understand, explain, and predict immunological phenomena. They help researchers study complex immune processes and responses that cannot be easily observed or manipulated in vivo.

Theoretical immunological models provide conceptual frameworks for understanding immune system behavior, often using diagrams or flowcharts to illustrate interactions between immune components. Mathematical models use mathematical equations to describe immune system dynamics, allowing researchers to simulate and analyze the outcomes of various scenarios. Computational models, also known as in silico models, are created using computer software and can incorporate both theoretical and mathematical concepts to create detailed simulations of immunological processes.

Immunological models are essential tools for advancing our understanding of the immune system and developing new therapies and vaccines. They enable researchers to test hypotheses, explore the implications of different assumptions, and identify areas requiring further investigation.

Cytosol refers to the liquid portion of the cytoplasm found within a eukaryotic cell, excluding the organelles and structures suspended in it. It is the site of various metabolic activities and contains a variety of ions, small molecules, and enzymes. The cytosol is where many biochemical reactions take place, including glycolysis, protein synthesis, and the regulation of cellular pH. It is also where some organelles, such as ribosomes and vesicles, are located. In contrast to the cytosol, the term "cytoplasm" refers to the entire contents of a cell, including both the cytosol and the organelles suspended within it.

Ferrosoferric oxide is commonly known as magnetite, which is a mineral form of iron(III) oxide (Fe2O3) and iron(II) oxide (FeO). Its chemical formula is often written as Fe3O4. It is a black colored, magnetic compound that occurs naturally in many environments, including rocks and soil. Magnetite has been used for various purposes throughout history, such as in the creation of early forms of magnetic storage media and as a pigment in paints. In the medical field, magnetite nanoparticles have been studied for potential use in targeted drug delivery systems and diagnostic imaging techniques.

Green Fluorescent Protein (GFP) is not a medical term per se, but a scientific term used in the field of molecular biology. GFP is a protein that exhibits bright green fluorescence when exposed to light, particularly blue or ultraviolet light. It was originally discovered in the jellyfish Aequorea victoria.

In medical and biological research, scientists often use recombinant DNA technology to introduce the gene for GFP into other organisms, including bacteria, plants, and animals, including humans. This allows them to track the expression and localization of specific genes or proteins of interest in living cells, tissues, or even whole organisms.

The ability to visualize specific cellular structures or processes in real-time has proven invaluable for a wide range of research areas, from studying the development and function of organs and organ systems to understanding the mechanisms of diseases and the effects of therapeutic interventions.

Growth inhibitors, in a medical context, refer to substances or agents that reduce or prevent the growth and proliferation of cells. They play an essential role in regulating normal cellular growth and can be used in medical treatments to control the excessive growth of unwanted cells, such as cancer cells.

There are two main types of growth inhibitors:

1. Endogenous growth inhibitors: These are naturally occurring molecules within the body that help regulate cell growth and division. Examples include retinoids, which are vitamin A derivatives, and interferons, which are signaling proteins released by host cells in response to viruses.

2. Exogenous growth inhibitors: These are synthetic or natural substances from outside the body that can be used to inhibit cell growth. Many chemotherapeutic agents and targeted therapies for cancer treatment fall into this category. They work by interfering with specific pathways involved in cell division, such as DNA replication or mitosis, or by inducing apoptosis (programmed cell death) in cancer cells.

It is important to note that growth inhibitors may also affect normal cells, which can lead to side effects during treatment. The challenge for medical researchers is to develop targeted therapies that specifically inhibit the growth of abnormal cells while minimizing harm to healthy cells.

Tuftsin is a tetrapeptide (a chain of four amino acids) that plays an important role in the immune system. It is derived from the Fc fragment of IgG (Immunoglobulin G) antibodies after they have interacted with antigens and been cleaved by proteolytic enzymes.

Tuftsin enhances phagocytosis, which is the process by which certain cells in the immune system engulf and destroy foreign substances, bacteria, or dead cells. Specifically, it activates and increases the mobility of neutrophils and monocytes/macrophages, two types of white blood cells involved in the immune response.

The sequence of amino acids in tuftsin is Thr-Lys-Pro-Arg (Threonine-Lysine-Proline-Arginine). The presence and activity of tuftsin are essential for optimal immune function, and deficiencies or abnormalities in tuftsin have been linked to various immunological disorders.

Gene expression regulation in bacteria refers to the complex cellular processes that control the production of proteins from specific genes. This regulation allows bacteria to adapt to changing environmental conditions and ensure the appropriate amount of protein is produced at the right time.

Bacteria have a variety of mechanisms for regulating gene expression, including:

1. Operon structure: Many bacterial genes are organized into operons, which are clusters of genes that are transcribed together as a single mRNA molecule. The expression of these genes can be coordinately regulated by controlling the transcription of the entire operon.
2. Promoter regulation: Transcription is initiated at promoter regions upstream of the gene or operon. Bacteria have regulatory proteins called sigma factors that bind to the promoter and recruit RNA polymerase, the enzyme responsible for transcribing DNA into RNA. The binding of sigma factors can be influenced by environmental signals, allowing for regulation of transcription.
3. Attenuation: Some operons have regulatory regions called attenuators that control transcription termination. These regions contain hairpin structures that can form in the mRNA and cause transcription to stop prematurely. The formation of these hairpins is influenced by the concentration of specific metabolites, allowing for regulation of gene expression based on the availability of those metabolites.
4. Riboswitches: Some bacterial mRNAs contain regulatory elements called riboswitches that bind small molecules directly. When a small molecule binds to the riboswitch, it changes conformation and affects transcription or translation of the associated gene.
5. CRISPR-Cas systems: Bacteria use CRISPR-Cas systems for adaptive immunity against viruses and plasmids. These systems incorporate short sequences from foreign DNA into their own genome, which can then be used to recognize and cleave similar sequences in invading genetic elements.

Overall, gene expression regulation in bacteria is a complex process that allows them to respond quickly and efficiently to changing environmental conditions. Understanding these regulatory mechanisms can provide insights into bacterial physiology and help inform strategies for controlling bacterial growth and behavior.

Esterases are a group of enzymes that catalyze the hydrolysis of ester bonds in esters, producing alcohols and carboxylic acids. They are widely distributed in plants, animals, and microorganisms and play important roles in various biological processes, such as metabolism, digestion, and detoxification.

Esterases can be classified into several types based on their substrate specificity, including carboxylesterases, cholinesterases, lipases, and phosphatases. These enzymes have different structures and mechanisms of action but all share the ability to hydrolyze esters.

Carboxylesterases are the most abundant and diverse group of esterases, with a wide range of substrate specificity. They play important roles in the metabolism of drugs, xenobiotics, and lipids. Cholinesterases, on the other hand, specifically hydrolyze choline esters, such as acetylcholine, which is an important neurotransmitter in the nervous system. Lipases are a type of esterase that preferentially hydrolyzes triglycerides and plays a crucial role in fat digestion and metabolism. Phosphatases are enzymes that remove phosphate groups from various molecules, including esters, and have important functions in signal transduction and other cellular processes.

Esterases can also be used in industrial applications, such as in the production of biodiesel, detergents, and food additives. They are often produced by microbial fermentation or extracted from plants and animals. The use of esterases in biotechnology is an active area of research, with potential applications in biofuel production, bioremediation, and medical diagnostics.

Medical Definition:

Superoxide dismutase (SOD) is an enzyme that catalyzes the dismutation of superoxide radicals (O2-) into oxygen (O2) and hydrogen peroxide (H2O2). This essential antioxidant defense mechanism helps protect the body's cells from damage caused by reactive oxygen species (ROS), which are produced during normal metabolic processes and can lead to oxidative stress when their levels become too high.

There are three main types of superoxide dismutase found in different cellular locations:
1. Copper-zinc superoxide dismutase (CuZnSOD or SOD1) - Present mainly in the cytoplasm of cells.
2. Manganese superoxide dismutase (MnSOD or SOD2) - Located within the mitochondrial matrix.
3. Extracellular superoxide dismutase (EcSOD or SOD3) - Found in the extracellular spaces, such as blood vessels and connective tissues.

Imbalances in SOD levels or activity have been linked to various pathological conditions, including neurodegenerative diseases, cancer, and aging-related disorders.

Ovalbumin is the major protein found in egg white, making up about 54-60% of its total protein content. It is a glycoprotein with a molecular weight of around 45 kDa and has both hydrophilic and hydrophobic regions. Ovalbumin is a single polypeptide chain consisting of 385 amino acids, including four disulfide bridges that contribute to its structure.

Ovalbumin is often used in research as a model antigen for studying immune responses and allergies. In its native form, ovalbumin is not allergenic; however, when it is denatured or degraded into smaller peptides through cooking or digestion, it can become an allergen for some individuals.

In addition to being a food allergen, ovalbumin has been used in various medical and research applications, such as vaccine development, immunological studies, and protein structure-function analysis.

Oligopeptides are defined in medicine and biochemistry as short chains of amino acids, typically containing fewer than 20 amino acid residues. These small peptides are important components in various biological processes, such as serving as signaling molecules, enzyme inhibitors, or structural elements in some proteins. They can be found naturally in foods and may also be synthesized for use in medical research and therapeutic applications.

Immune evasion is a term used in immunology to describe the various strategies employed by pathogens (such as viruses, bacteria, parasites) to avoid or subvert the host's immune system. This can include mechanisms that allow the pathogen to directly inhibit or escape the actions of immune cells, like T cells and neutrophils, or to prevent the detection of their presence by masking themselves from the immune system.

For example, some viruses may change their surface proteins to avoid recognition by antibodies, while others may block the presentation of their antigens to T cells. Similarly, some bacteria can produce enzymes that degrade or modify components of the immune system, allowing them to evade detection and destruction.

Immune evasion is a major challenge in the development of effective vaccines and therapies for infectious diseases, as it allows pathogens to persist and cause chronic infections. Understanding the mechanisms of immune evasion can help researchers develop strategies to overcome these challenges and improve outcomes for patients.

A primary cell culture is the very first cell culture generation that is established by directly isolating cells from an original tissue or organ source. These cells are removed from the body and then cultured in controlled conditions in a laboratory setting, allowing them to grow and multiply. Primary cell cultures maintain many of the characteristics of the cells in their original tissue environment, making them valuable for research purposes. However, they can only be passaged (subcultured) a limited number of times before they undergo senescence or change into a different type of cell.

The tumor microenvironment (TME) is a complex and dynamic setting that consists of various cellular and non-cellular components, which interact with each other and contribute to the growth, progression, and dissemination of cancer. The TME includes:

1. Cancer cells: These are the malignant cells that grow uncontrollably, invade surrounding tissues, and can spread to distant organs.
2. Stromal cells: These are non-cancerous cells present within the tumor, including fibroblasts, immune cells, adipocytes, and endothelial cells. They play a crucial role in supporting the growth of cancer cells by providing structural and nutritional support, modulating the immune response, and promoting angiogenesis (the formation of new blood vessels).
3. Extracellular matrix (ECM): This is the non-cellular component of the TME, consisting of a network of proteins, glycoproteins, and polysaccharides that provide structural support and regulate cell behavior. The ECM can be remodeled by both cancer and stromal cells, leading to changes in tissue stiffness, architecture, and signaling pathways.
4. Soluble factors: These include various cytokines, chemokines, growth factors, and metabolites that are secreted by both cancer and stromal cells. They can act as signaling molecules, influencing cell behavior, survival, proliferation, and migration.
5. Blood vessels: The formation of new blood vessels (angiogenesis) within the TME is essential for providing nutrients and oxygen to support the growth of cancer cells. The vasculature in the TME is often disorganized, leading to hypoxic (low oxygen) regions and altered drug delivery.
6. Immune cells: The TME contains various immune cell populations, such as tumor-associated macrophages (TAMs), dendritic cells, natural killer (NK) cells, and different subsets of T lymphocytes. These cells can either promote or inhibit the growth and progression of cancer, depending on their phenotype and activation status.
7. Niche: A specific microenvironment within the TME that supports the survival and function of cancer stem cells (CSCs) or tumor-initiating cells. The niche is often characterized by unique cellular components, signaling molecules, and physical properties that contribute to the maintenance and propagation of CSCs.

Understanding the complex interactions between these various components in the TME can provide valuable insights into cancer biology and help inform the development of novel therapeutic strategies.

Cytochalasin B is a fungal metabolite that inhibits actin polymerization in cells, which can disrupt the cytoskeleton and affect various cellular processes such as cell division and motility. It is often used in research to study actin dynamics and cell shape.

Immunization is defined medically as the process where an individual is made immune or resistant to an infectious disease, typically through the administration of a vaccine. The vaccine stimulates the body's own immune system to recognize and fight off the specific disease-causing organism, thereby preventing or reducing the severity of future infections with that organism.

Immunization can be achieved actively, where the person is given a vaccine to trigger an immune response, or passively, where antibodies are transferred to the person through immunoglobulin therapy. Immunizations are an important part of preventive healthcare and have been successful in controlling and eliminating many infectious diseases worldwide.

Interferon Regulatory Factor-1 (IRF-1) is a protein that belongs to the Interferon Regulatory Factor family. It functions as a transcription factor, which means it regulates the expression of specific genes. IRF-1 plays a crucial role in regulating the immune response and inflammation.

More specifically, IRF-1 is involved in the signaling pathways that are activated by interferons (IFNs), which are proteins released by cells in response to viral or bacterial infections. Once activated, IRF-1 binds to specific DNA sequences in the promoter regions of target genes and activates their transcription.

IRF-1 regulates the expression of a variety of genes involved in the immune response, including those that encode cytokines, chemokines, and major histocompatibility complex (MHC) molecules. It also plays a role in the regulation of cell growth, differentiation, and apoptosis (programmed cell death).

Mutations or dysregulation of IRF-1 have been implicated in various diseases, including cancer, autoimmune disorders, and viral infections.

Oxygen is a colorless, odorless, tasteless gas that constitutes about 21% of the earth's atmosphere. It is a crucial element for human and most living organisms as it is vital for respiration. Inhaled oxygen enters the lungs and binds to hemoglobin in red blood cells, which carries it to tissues throughout the body where it is used to convert nutrients into energy and carbon dioxide, a waste product that is exhaled.

Medically, supplemental oxygen therapy may be provided to patients with conditions such as chronic obstructive pulmonary disease (COPD), pneumonia, heart failure, or other medical conditions that impair the body's ability to extract sufficient oxygen from the air. Oxygen can be administered through various devices, including nasal cannulas, face masks, and ventilators.

Cathepsin B is a lysosomal cysteine protease that plays a role in various physiological processes, including intracellular protein degradation, antigen presentation, and extracellular matrix remodeling. It is produced as an inactive precursor (procathepsin B) and activated upon cleavage of the propeptide by other proteases or autocatalytically. Cathepsin B has a wide range of substrates, including collagen, elastin, and various intracellular proteins. Its dysregulation has been implicated in several pathological conditions, such as cancer, neurodegenerative diseases, and inflammatory disorders.

"Competitive binding" is a term used in pharmacology and biochemistry to describe the behavior of two or more molecules (ligands) competing for the same binding site on a target protein or receptor. In this context, "binding" refers to the physical interaction between a ligand and its target.

When a ligand binds to a receptor, it can alter the receptor's function, either activating or inhibiting it. If multiple ligands compete for the same binding site, they will compete to bind to the receptor. The ability of each ligand to bind to the receptor is influenced by its affinity for the receptor, which is a measure of how strongly and specifically the ligand binds to the receptor.

In competitive binding, if one ligand is present in high concentrations, it can prevent other ligands with lower affinity from binding to the receptor. This is because the higher-affinity ligand will have a greater probability of occupying the binding site and blocking access to the other ligands. The competition between ligands can be described mathematically using equations such as the Langmuir isotherm, which describes the relationship between the concentration of ligand and the fraction of receptors that are occupied by the ligand.

Competitive binding is an important concept in drug development, as it can be used to predict how different drugs will interact with their targets and how they may affect each other's activity. By understanding the competitive binding properties of a drug, researchers can optimize its dosage and delivery to maximize its therapeutic effect while minimizing unwanted side effects.

STAT6 (Signal Transducer and Activator of Transcription 6) is a transcription factor that plays a crucial role in the immune response, particularly in the development of Th2 cells and the production of cytokines. It is activated by cytokines such as IL-4 and IL-13 through phosphorylation, which leads to its dimerization and translocation into the nucleus where it binds to specific DNA sequences and regulates the expression of target genes. STAT6 is involved in a variety of biological processes including allergic responses, inflammation, and tumorigenesis. Mutations in the STAT6 gene have been associated with immunodeficiency disorders and certain types of cancer.

Experimental arthritis refers to the induction of joint inflammation in animal models for the purpose of studying the disease process and testing potential treatments. This is typically achieved through the use of various methods such as injecting certain chemicals or proteins into the joints, genetically modifying animals to develop arthritis-like symptoms, or immunizing animals to induce an autoimmune response against their own joint tissues. These models are crucial for advancing our understanding of the underlying mechanisms of arthritis and for developing new therapies to treat this debilitating disease.

C-X-C chemokine receptor type 4 (CXCR4) is a type of protein found on the surface of some cells, including white blood cells, and is a type of G protein-coupled receptor (GPCR). CXCR4 binds specifically to the chemokine ligand CXCL12 (also known as stromal cell-derived factor 1, or SDF-1), which plays a crucial role in the trafficking and homing of immune cells, particularly hematopoietic stem cells and lymphocytes. The binding of CXCL12 to CXCR4 triggers various intracellular signaling pathways that regulate cell migration, proliferation, survival, and differentiation.

In addition to its role in the immune system, CXCR4 has been implicated in several physiological and pathological processes, such as embryonic development, neurogenesis, angiogenesis, cancer metastasis, and HIV infection. In cancer, the overexpression of CXCR4 or increased levels of its ligand CXCL12 have been associated with poor prognosis, tumor growth, and metastasis in various types of malignancies, including breast, lung, prostate, colon, and ovarian cancers. In HIV infection, the CXCR4 coreceptor, together with CD4, facilitates viral entry into host cells, particularly during the later stages of the disease when the virus shifts its preference from CCR5 to CXCR4 as a coreceptor.

In summary, CXCR4 is a cell-surface receptor that binds specifically to the chemokine ligand CXCL12 and plays essential roles in immune cell trafficking, hematopoiesis, cancer metastasis, and HIV infection.

I'm sorry for any confusion, but "Sarcoma, Experimental" is not a recognized medical term or definition. Sarcomas are a type of cancer that develop in the body's connective tissues, such as bones, muscles, tendons, cartilage, and fat. There are many different types of sarcomas, classified based on the specific type of tissue they originate from.

Experimental, on the other hand, refers to something that is being tested or tried out for the first time, typically as part of a scientific experiment or clinical trial. In the context of cancer treatment, an experimental therapy might refer to a new drug, procedure, or device that is still being studied in clinical trials to determine its safety and effectiveness.

Therefore, "Sarcoma, Experimental" could potentially refer to a clinical trial or research study involving a new treatment for sarcoma, but it would not be a medical definition in and of itself. If you have any specific questions about sarcomas or experimental treatments, I would recommend consulting with a healthcare professional or medical researcher for more accurate information.

Calgranulin A is also known as S100A8 or MRP-14. It is a calcium-binding protein that belongs to the S100 family of proteins. Calgranulin A is primarily found in the cytoplasm of neutrophils, a type of white blood cell involved in inflammation and immune response.

Calgranulin A can be released from neutrophils during inflammation and has been implicated in various biological processes, including regulation of innate immunity, inflammation, and cancer progression. It can also interact with other proteins to form heterodimers or multimers, such as calprotectin (S100A8/S100A9), which has been associated with several pathological conditions, including autoimmune diseases, infections, and cancer.

In medical research, Calgranulin A is often used as a biomarker for various inflammatory conditions, such as rheumatoid arthritis, inflammatory bowel disease, and chronic obstructive pulmonary disease (COPD). Elevated levels of Calgranulin A in body fluids, such as blood or sputum, may indicate the presence of an ongoing inflammatory response.

I'm assuming you are asking for information about "Ly" antigens in the context of human immune system and immunology.

Ly (Lymphocyte) antigens are a group of cell surface markers found on human leukocytes, including T cells, NK cells, and some B cells. These antigens were originally identified through serological analysis and were historically used to distinguish different subsets of lymphocytes based on their surface phenotype.

The "Ly" nomenclature has been largely replaced by the CD (Cluster of Differentiation) system, which is a more standardized and internationally recognized classification system for cell surface markers. However, some Ly antigens are still commonly referred to by their historical names, such as:

* Ly-1 or CD5: A marker found on mature T cells, including both CD4+ and CD8+ subsets.
* Ly-2 or CD8: A marker found on cytotoxic T cells, which are a subset of CD8+ T cells that can directly kill infected or damaged cells.
* Ly-3 or CD56: A marker found on natural killer (NK) cells, which are a type of immune cell that can recognize and destroy virus-infected or cancerous cells without the need for prior activation.

It's worth noting that while these antigens were originally identified through serological analysis, they are now more commonly detected using flow cytometry, which allows for the simultaneous measurement of multiple surface markers on individual cells. This has greatly expanded our ability to identify and characterize different subsets of immune cells and has led to a better understanding of their roles in health and disease.

Medical Definition of Monoglycerides:

Monoglycerides are types of glycerides that contain one molecule of fatty acid combined with a glycerol molecule through an ester linkage. They are often used as food additives, serving as emulsifiers to help blend together water and oil-based ingredients in foods such as baked goods, ice cream, and chocolate. Monoglycerides can also be found naturally in some foods, including certain vegetable oils.

In the context of human physiology, monoglycerides can serve as a source of energy and can also play a role in the absorption and transport of fatty acids in the body. However, they are not typically considered to be a major nutrient or component of the human diet.

Calgranulin B is also known as S100 calcium-binding protein B or S100A9. It is a calcium-binding protein that plays a role in inflammation and immune response. Calgranulin B can be found in granulocytes, monocytes, and some epithelial cells. It forms heterocomplexes with calgranulin A (S100A8) and these complexes are involved in the regulation of innate immunity and inflammation. They can act as damage-associated molecular patterns (DAMPs) and contribute to the pathogenesis of various inflammatory diseases, such as rheumatoid arthritis, inflammatory bowel disease, and certain types of cancer.

Tumor Necrosis Factor (TNF) Receptors are cell surface receptors that bind to tumor necrosis factor cytokines. They play crucial roles in the regulation of a variety of immune cell functions, including inflammation, immunity, and cell survival or death (apoptosis).

There are two major types of TNF receptors: TNFR1 (also known as p55 or CD120a) and TNFR2 (also known as p75 or CD120b). TNFR1 is widely expressed in most tissues, while TNFR2 has a more restricted expression pattern and is mainly found on immune cells.

TNF receptors have an intracellular domain called the death domain, which can trigger signaling pathways leading to apoptosis when activated by TNF ligands. However, they can also activate other signaling pathways that promote cell survival, differentiation, and inflammation. Dysregulation of TNF receptor signaling has been implicated in various diseases, including cancer, autoimmune disorders, and neurodegenerative conditions.

Tuberculin is not a medical condition but a diagnostic tool used in the form of a purified protein derivative (PPD) to detect tuberculosis infection. It is prepared from the culture filtrate of Mycobacterium tuberculosis, the bacterium that causes TB. The PPD tuberculin is injected intradermally, and the resulting skin reaction is measured after 48-72 hours to determine if a person has developed an immune response to the bacteria, indicating a past or present infection with TB. It's important to note that a positive tuberculin test does not necessarily mean that active disease is present, but it does indicate that further evaluation is needed.

An animal model in medicine refers to the use of non-human animals in experiments to understand, predict, and test responses and effects of various biological and chemical interactions that may also occur in humans. These models are used when studying complex systems or processes that cannot be easily replicated or studied in human subjects, such as genetic manipulation or exposure to harmful substances. The choice of animal model depends on the specific research question being asked and the similarities between the animal's and human's biological and physiological responses. Examples of commonly used animal models include mice, rats, rabbits, guinea pigs, and non-human primates.

Culture techniques are methods used in microbiology to grow and multiply microorganisms, such as bacteria, fungi, or viruses, in a controlled laboratory environment. These techniques allow for the isolation, identification, and study of specific microorganisms, which is essential for diagnostic purposes, research, and development of medical treatments.

The most common culture technique involves inoculating a sterile growth medium with a sample suspected to contain microorganisms. The growth medium can be solid or liquid and contains nutrients that support the growth of the microorganisms. Common solid growth media include agar plates, while liquid growth media are used for broth cultures.

Once inoculated, the growth medium is incubated at a temperature that favors the growth of the microorganisms being studied. During incubation, the microorganisms multiply and form visible colonies on the solid growth medium or turbid growth in the liquid growth medium. The size, shape, color, and other characteristics of the colonies can provide important clues about the identity of the microorganism.

Other culture techniques include selective and differential media, which are designed to inhibit the growth of certain types of microorganisms while promoting the growth of others, allowing for the isolation and identification of specific pathogens. Enrichment cultures involve adding specific nutrients or factors to a sample to promote the growth of a particular type of microorganism.

Overall, culture techniques are essential tools in microbiology and play a critical role in medical diagnostics, research, and public health.

"Mycobacterium leprae" is a slow-growing, rod-shaped, gram-positive bacterium that is the causative agent of leprosy, a chronic infectious disease that primarily affects the skin, peripheral nerves, and mucosal surfaces of the upper respiratory tract. The bacterium was discovered in 1873 by Gerhard Armauer Hansen, a Norwegian physician, and is named after him as "Hansen's bacillus."

"Mycobacterium leprae" has a unique cell wall that contains high amounts of lipids, which makes it resistant to many common disinfectants and antibiotics. It can survive and multiply within host macrophages, allowing it to evade the immune system and establish a chronic infection.

Leprosy is a treatable disease with multidrug therapy (MDT), which combines several antibiotics such as dapsone, rifampicin, and clofazimine. Early diagnosis and treatment can prevent the progression of the disease and reduce its transmission to others.

The extracellular matrix (ECM) is a complex network of biomolecules that provides structural and biochemical support to cells in tissues and organs. It is composed of various proteins, glycoproteins, and polysaccharides, such as collagens, elastin, fibronectin, laminin, and proteoglycans. The ECM plays crucial roles in maintaining tissue architecture, regulating cell behavior, and facilitating communication between cells. It provides a scaffold for cell attachment, migration, and differentiation, and helps to maintain the structural integrity of tissues by resisting mechanical stresses. Additionally, the ECM contains various growth factors, cytokines, and chemokines that can influence cellular processes such as proliferation, survival, and differentiation. Overall, the extracellular matrix is essential for the normal functioning of tissues and organs, and its dysregulation can contribute to various pathological conditions, including fibrosis, cancer, and degenerative diseases.

Therapeutic irrigation, also known as lavage, is a medical procedure that involves the introduction of fluids or other agents into a body cavity or natural passageway for therapeutic purposes. This technique is used to cleanse, flush out, or introduce medication into various parts of the body, such as the bladder, lungs, stomach, or colon.

The fluid used in therapeutic irrigation can be sterile saline solution, distilled water, or a medicated solution, depending on the specific purpose of the procedure. The flow and pressure of the fluid are carefully controlled to ensure that it reaches the desired area without causing damage to surrounding tissues.

Therapeutic irrigation is used to treat a variety of medical conditions, including infections, inflammation, obstructions, and toxic exposures. It can also be used as a diagnostic tool to help identify abnormalities or lesions within body cavities.

Overall, therapeutic irrigation is a valuable technique in modern medicine that allows healthcare providers to deliver targeted treatment directly to specific areas of the body, improving patient outcomes and quality of life.

... (Mregs) represent a subset of anti-inflammatory macrophages. In general, macrophages are a very dynamic ... Coculture of macrophages with regulatory T cells (Tregs) caused differentiation of macrophages toward Mreg phenotype. Similar ... M2a, mainly known as alternatively activated macrophages, are macrophages associated with tissue healing due to the production ... Surprisingly, Mregs resemble classically activated macrophages more than alternatively activated macrophages, due to higher ...
M1 macrophage demonstrates high TF, HAMP, and FTH1 gene activities that mediates iron uptake. M2 macrophage on the other hand ... The reprogram of macrophage implies a dysregulated inflammation that can damage healthy lung cells. Macrophage polarization is ... The presence of these dirty macrophages has been a characteristic of many smoke-related lung diseases. A macrophage can be ... Some of the smoker's macrophages are recruited from the circulating monocytes while some are the original alveolar macrophages ...
For example, macrophages stimulated with IL-4 and IL-13 are defined as M2a, whereas macrophages stimulated with LPS and ... Macrophages displaying M1 phenotype have been characterized by expression of F4/80, CD11c and iNOS whereas macrophages ... Adipose tissue macrophages (abbr. ATMs) comprise tissue resident macrophages present in adipose tissue. Adipose tissue apart ... M1 macrophages are microbicidal and tumoricidal, and stimulate adaptive immune response. M2 macrophages are associated with ...
... and the activated macrophages are known as classically activated macrophages, or M1 macrophages. The M1 macrophages in turn ... or M1 macrophages, and the alternatively activated macrophages, or M2 macrophages. M1 macrophages are proinflammatory, while M2 ... which activate macrophages to become M2 macrophages, also known as alternatively activated macrophages. M2 macrophages express ... M1 macrophages: as mentioned earlier (previously referred to as classically activated macrophages), M1 "killer" macrophages are ...
Skin Hair follicle Macrophage Dendritic cell Langerhans cell Macrophage polarisation Inflammation Phenotypes of macrophages ... Numerous dermal macrophages are present in the skin at birth due to the infiltration of yolk-sac derived macrophages and ... Dermal macrophages are macrophages in the skin that facilitate skin homeostasis by mediating wound repair, hair growth, and ... M1 macrophages can suppress tumour growth in the skin by their pro-inflammatory properties. However, M2 macrophages support ...
... (EC 3.4.24.65, metalloelastase, human macrophage metalloelastase (HME), MMP-12) is an enzyme. This enzyme ... Macrophage+elastase at the U.S. National Library of Medicine Medical Subject Headings (MeSH) Portal: Biology (EC 3.4.24). ... Kettner C, Shaw E, White R, Janoff A (May 1981). "The specificity of macrophage elastase on the insulin B-chain". The ... Matrix metallopeptidase 12 Banda MJ, Werb Z (February 1981). "Mouse macrophage elastase. Purification and characterization as a ...
The diversity of macrophage phenotypes still remain to be fully characterized in vivo. The imbalance of the macrophage types is ... Macrophage polarization is a process by which macrophages adopt different functional programs in response to the signals from ... This plasticity of macrophage phenotype has added to the confusion regarding the existence of individual macrophage sub-types ... By simplified classification, macrophage phenotype has been divided into 2 groups: M1 (classically activated macrophages) and ...
The terms "macrophage" and "microphage" are used in ecology to describe heterotrophs that consume food in two different ways. ... Both macrophages and microphages "ingest solid food and may process it through some sort of alimentary canal." However, a ... The terms "macrophage" and "microphage" were originally used in this sense by Jordan and Hirsch (1927; cited in Yonge 1928). ... Although they have been used in ecology texts as recently as 2002, the terms macrophage and microphage today are primarily used ...
An alveolar macrophage, pulmonary macrophage, (or dust cell) is a type of macrophage, a professional phagocyte, found in the ... The alveolar macrophage is the third cell type in the alveolus; the others are the type I and type II pneumocytes. Alveolar ... Alveolar macrophages are also involved in the phagocytosis of apoptotic and necrotic cells. They need to be selective of the ... Alveolar macrophages are frequently seen to contain granules of exogenous material such as particulate carbon that they have ...
The phrase 'alternatively activated macrophage' is used to refer to M2 macrophages. Regulatory macrophages do not fit into the ... Analysis of the macrophage population and signaling in a tumor may provide useful clinical data. Macrophages have been ... A macrophage-activating factor (MAF) is a lymphokine or other receptor based signal that primes macrophages towards ... Macrophages inherently display tissue and environment-dependent plasticity. In addition, the phenotypes of the macrophages in a ...
A tingible body macrophage (TBMs) is a type of macrophage predominantly found in germinal centers, containing many phagocytized ... v t e (Articles with short description, Short description matches Wikidata, Macrophages, All stub articles, Immunology stubs). ... Smith JP, Burton GF, Tew JG, Szakal AK (1998). "Tingible body macrophages in regulation of germinal center reactions". ... Horst Ibelgaufts' COPE: Cytokines & Cells Online Pathfinder Encyclopaedia > tingible body macrophages Retrieved on June 27, ...
Islet resident macrophages are the predominant myeloid cell of the pancreatic islets of langerhans. Islet resident macrophages ... showing that islet resident macrophages promote beta cell proliferation suggest that islet macrophages are likely important ... Islet resident macrophages depend on the expression of colony stimulating factor (CSF1). Knockout mice for the CSF-1 gene cause ... As opposed to macrophages in the islet stroma (tissue), whose developmental origin is from the yolk sac (primitive ...
Macrophages are known to originate from bone marrow-derived blood monocytes (monocyte-derived macrophages) or yolk sac ... The composition of monocyte-derived macrophages and tissue-resident macrophages in the tumor microenvironment depends on the ... M1 macrophages are seen to have a pro-inflammatory and cytotoxic (anti-tumoral) function; M2 macrophages are anti-inflammatory ... quickly turn into tumor-associated macrophages. Additionally, the crosstalk between M-MDSCs and other macrophages enhance the ...
... (CAPG) also known as actin regulatory protein CAP-G is a protein that in humans is encoded by the ... Dabiri GA, Young CL, Rosenbloom J, Southwick FS (Sep 1992). "Molecular cloning of human macrophage capping protein cDNA. A ... Southwick FS (1995). "Gain-of-function mutations conferring actin-severing activity to human macrophage cap G". J. Biol. Chem. ... unique member of the gelsolin/villin family expressed primarily in macrophages". J Biol Chem. 267 (23): 16545-52. doi:10.1016/ ...
A febrile patient with known or suspected SoJIA must be considered for macrophage activation if: Ferritin >684 ng/ml and any 2 ... Macrophage activation syndrome is a severe, potentially life-threatening, complication of several chronic rheumatic diseases of ... There is uncontrolled activation and proliferation of macrophages, and T lymphocytes, with a marked increase in circulating ...
Other macrophage inflammatory proteins include MIP-2, MIP-3 and MIP-5. MIP-1α and MIP-1β are major factors produced by ... Macrophage Inflammatory Proteins (MIP) belong to the family of chemotactic cytokines known as chemokines. In humans, there are ... Macrophage+Inflammatory+Proteins at the U.S. National Library of Medicine Medical Subject Headings (MeSH) (Articles with short ... ISBN 978-0-8153-4505-3. Qin CC, Liu YN, Hu Y, Yang Y, Chen Z (May 2017). "Macrophage inflammatory protein-2 as mediator of ...
... (or integrin αMβ2 or macrophage integrin or Mac-1) is a complement receptor ("CR3") consisting of CD11b ( ... CR3 CD11b/CD18 Macrophage 1 antigen (Mac-1) Macrophage Todd R (1996). "The continuing saga of complement receptor type 3 (CR3 ... Macrophage-1 antigen (hereafter complement receptor 3 or CR3) (CD11b/CD18) is a human cell surface receptor found on B and T ... Macrophage-1+antigen at the U.S. National Library of Medicine Medical Subject Headings (MeSH) (CS1 maint: location missing ...
... macrophages, and bone marrow progenitor cells. M-CSF affects macrophages and monocytes in several ways, including stimulating ... Macrophage+Colony-Stimulating+Factor at the U.S. National Library of Medicine Medical Subject Headings (MeSH) Overview of all ... The role of M-CSF is not only restricted to the monocyte/macrophage cell lineage. By interacting with its membrane receptor ( ... "Entrez Gene: CSF1 colony stimulating factor 1 (macrophage)". Jang MH, Herber DM, Jiang X, Nandi S, Dai XM, Zeller G, Stanley ER ...
Bone-marrow-derived macrophage (BMDM) refers to macrophage cells that are generated in a research laboratory from mammalian ... BMDMs provide a large homogenous population of macrophages that play an increasingly important role in making macrophage- ... which then mature into macrophages). Macrophages have a wide variety of functions in the body including phagocytosis of foreign ... the precursor cells to macrophages, which are less helpful for macrophage-specific research Barrett JP, Costello DA, O'Sullivan ...
The lipid-laden macrophage index (LLMI) can be calculated by counting 100 macrophages in a BAL specimen treated with a lipid ... specimens that consist of macrophages containing deposits of lipids (fats). The lipid content of the macrophages can be ... Lipid-laden alveolar macrophages, also known as pulmonary foam cells, are cells found in bronchoalveolar lavage (BAL) ... Lipid-laden alveolar macrophages have been reported in cases of vaping-associated pulmonary injury. ...
... has been reported to interact with: BNIPL, CD74, COPS5, CXCR4, and RPS19. MIF is a ... Macrophage migration inhibitory factor assembles into a trimer composed of three identical subunits. Each of these monomers ... Media related to Macrophage migration inhibitory factor at Wikimedia Commons (CS1 Chinese-language sources (zh), Articles with ... Wang F, Shen X, Guo X, Peng Y, Liu Y, Xu S, Yang J (February 2010). "Spinal macrophage migration inhibitory factor contributes ...
... is an evolutionary conserved protein domain. Macrophage migration inhibitory ... MIF is released from T-cells and macrophages, and acts within the neuroendocrine system. MIF is capable of tautomerase activity ... Matsunaga J, Sinha D, Solano F, Santis C, Wistow G, Hearing V (November 1999). "Macrophage migration inhibitory factor (MIF)-- ... Donn RP, Ray DW (July 2004). "Macrophage migration inhibitory factor: molecular, cellular and genetic aspects of a key ...
... s (Nramps), also known as metal ion (Mn2+-iron) transporters (TC# 2.A.55), are ... In contrast to the widely expressed NRAMP2, NRAMP1 is expressed primarily in macrophages and monocytes and appears to have a ... The name "natural resistance-associated" macrophage proteins arises from the role in resistance of intracellular bacterial ... for Mn2+ or some other metal prevents the generation of reactive oxygenic and nitrogenic compounds that are used by macrophage ...
In macrophages, it has also been shown to signal via STAT3. The cytokine activates macrophages to inhibit fungal survival. It ... "Granulocyte macrophage-colony stimulating factor induced Zn sequestration enhances macrophage superoxide and limits ... Human granulocyte-macrophage colony-stimulating factor is glycosylated in its mature form.[citation needed] GM-CSF was first ... Granulocyte-macrophage colony-stimulating factor (GM-CSF), also known as colony-stimulating factor 2 (CSF2), is a monomeric ...
... (NRAMP 2), also known as divalent metal transporter 1 (DMT1) and divalent ... Kishi F, Tabuchi M (October 1998). "Human natural resistance-associated macrophage protein 2: gene cloning and protein ...
... is a protein that in humans is encoded by the SLC11A1 gene. This gene is a ... Søborg C, Andersen AB, Madsen HO, Kok-Jensen A, Skinhøj P, Garred P (August 2002). "Natural resistance-associated macrophage ... Kishi F, Nobumoto M (1996). "Identification of natural resistance-associated macrophage protein in peripheral blood lymphocytes ... "Nramp1 locus encodes a 65 kDa interferon-gamma-inducible protein in murine macrophages". The Biochemical Journal. 325. 325 ( Pt ...
The granulocyte-macrophage colony-stimulating factor receptor also known as CD116 (Cluster of Differentiation 116), is a ... Remaining extracellular domain acts as a soluble GM-CSFRα and have been identified in bone marrow, monocytes and macrophages, ... Gearing DP, King JA, Gough NM, Nicola NA (Dec 1989). "Expression cloning of a receptor for human granulocyte-macrophage colony- ... Lia F, Rajotte D, Clark SC, Hoang T (Nov 1996). "A dominant negative granulocyte-macrophage colony-stimulating factor receptor ...
"Macrophages". British Society for Immunology. Retrieved 8 November 2018. Annunziato, F; Romagnani, C; Romagnani, S (March 2015 ... By secreting interferon gamma and TNF, TH1, TC1, and group 1 ILCS activate macrophages, converting them to potent effector ... Type 1 immunity is directed primarily at viruses, bacteria, and protozoa and is responsible for activating macrophages, turning ... for macrophages); and Stimulating cells to secrete a variety of cytokines that influence the function of other cells involved ...
Macrophages can be stimulated by T cell secretion of interferon. After this activation, macrophages are able to express MHC ... "Macrophages". Access Science. 2015. doi:10.1036/1097-8542.BR1109151. Dalod M, Chelbi R, Malissen B, Lawrence T (May 2014). " ... At the affected site, the macrophage surrounds the site of infection or tissue damage with its membrane in a mechanism called ... The main types of professional antigen-presenting cells are dendritic cells, macrophages and B cells. Dendritic cells have the ...
Macrophages are part of the human immune system. They are involved in the detection, phagocytosis and destruction of organisms ... "Macrophages , British Society for Immunology". www.immunology.org. Retrieved 2022-12-15. Saman, S.; Stagno, M. J.; Warmann, S. ... It is a non-invasive form of liquid biopsy, i.e. biopsy from blood, which analyzes activated macrophages (CD14+/CD16+) for ... In case of cancerous tumors, macrophages ingest tumor cells and dissolve them with the help of enzymes, storing tumor proteins ...
Regulatory macrophages (Mregs) represent a subset of anti-inflammatory macrophages. In general, macrophages are a very dynamic ... Coculture of macrophages with regulatory T cells (Tregs) caused differentiation of macrophages toward Mreg phenotype. Similar ... M2a, mainly known as alternatively activated macrophages, are macrophages associated with tissue healing due to the production ... Surprisingly, Mregs resemble classically activated macrophages more than alternatively activated macrophages, due to higher ...
Macrophages Drive Regeneration. The activity of one type of immune cell helps regrow the limbs of amputated salamanders.. Edyta ... Macrophages are known to be important in human muscle repair after acute damage. To test their role in limb regeneration in the ... AxolotlWIKIMEDIA, STAN SHEBSMacrophages, a type of immune cell that travels throughout tissues gobbling up bacteria and dead ... axolotl, researchers from the Australian Regenerative Medicine Institute depleted macrophages in amputated salamanders and ...
Macrophage activation syndrome is characterized by pancytopenia, liver insufficiency,... ... Macrophage activation syndrome (MAS) is a life-threatening complication of rheumatic disease that, for unknown reasons, occurs ... encoded search term (Macrophage Activation Syndrome) and Macrophage Activation Syndrome What to Read Next on Medscape ... Macrophage activation syndrome in rheumatic diseases in children]. Rev Rhum Ed Fr. 1994 Jun. 61(6):447-51. [QxMD MEDLINE Link] ...
The presence of immune cells, such as macrophages, in metabolic tissues suggests dynamic, ongoing crosstalk between these two ... Here, we discuss how changes in the recruitment and activation of macrophages contribute … ... Macrophage-mediated inflammation in metabolic disease Nat Rev Immunol. 2011 Oct 10;11(11):738-49. doi: 10.1038/nri3071. ... The presence of immune cells, such as macrophages, in metabolic tissues suggests dynamic, ongoing crosstalk between these two ...
Whether the sequential presence of two different macrophage populations results from a dynamic shift in macrophage polarization ... Perturbations of macrophage function and/or activation may result in impaired regeneration and fibrosis deposition as described ... A lot of evidence demonstrated that after acute injury M1 macrophages infiltrate early to promote the clearance of necrotic ... In this paper, we discuss the current available information about the role that different phenotypes of macrophages plays after ...
In addition, a new mechanism of inflammatory control of macrophage diversity based on lactate-mediated histone modifications ... A lactate timer that controls inflammatory responses in macrophages was also described. ... single-cell sequencing combined with new macrophage-specific genetic tools has provided unexpected insights into the tissue- ... single-cell sequencing studies provided important insight into the diverse gene expression profiles of tissue macrophages, and ...
Macrophage definition: Any of the large phagocytic cells of the reticuloendothelial system. ... Macrophage Sentence Examples *. Because the bacteria live within macrophage cells, treatment may be difficult, and the ... The cells were collected after the daughter was given granulocyte macrophage colony stimulating factor for 5 days to boost her ... However, we have now extended these studies to use retroviral gene transfer methods to stably transduce macrophage precursors ...
Microglia are similar, but not identical to other tissue macrophages, and in this review, we will first summarize the ... Then, we will discuss several principles that govern macrophage phagocytosis of apoptotic cells (efferocytosis), including the ... Then, we will discuss several principles that govern macrophage phagocytosis of apoptotic cells (efferocytosis), including the ... lineage and population maintenance of microglia and macrophages. ... lineage and population maintenance of microglia and macrophages ...
Macrophage - A type of white blood cell that surrounds and kills microorganisms, removes dead cells, and stimulates the action ... Medical Word - Macrophage. Ans : A type of white blood cell that surrounds and kills microorganisms, removes dead cells, and ... Macrophage - Glossary. Written & Compiled by Medindia Content Team. Medically Reviewed by The Medindia Medical Review Team on ...
Macrophages, as one of the most abundant tumor-infiltrating cells, play an important role in tumor development and metastasis. ... Pro-inflammatory M1 macrophages hold the potential to engulf tumor cells. In contrast, anti-inflammatory M2 macrophages, which ... Herein, we introduce the role of macrophages in tumor progression, summarize the recent advances in macrophage-centered ... Polarization profiles of human M-CSF-generated macrophages and comparison of M1-markers in classically activated macrophages ...
We suggest macrophage-specific targeting of the CD44v7 pathway as a novel therapeutic option for Crohns disease. ... Importantly, stimulation of T cells by Cd44v7-deficient macrophages induced upregulation of Foxp3 in vitro, while cotransfer of ... In contrast, CD44v7 absence on macrophages in recipient mice prevented colitis. Prevention was associated with the ... Cd44WT macrophages into Cd44v7-deficient mice reduced Foxp3+ T-cell counts and caused colitis. Accordingly, the CD44v7 ligand ...
... the current knowledge on the many functions of autophagy proteins in cell defenses with a focus on bacteria-macrophage ... the current knowledge on the many functions of autophagy proteins in cell defenses with a focus on bacteria-macrophage ... Autophagy evasion strategies adopted by bacterial pathogens inside macrophages. To evade autophagy in macrophages bacterial ... ATG16L1 deficiency in macrophages drives clearance of uropathogenic E. coli in an IL-1beta-dependent manner. Mucosal Immunol ( ...
New cell culture model for human alveolar macrophages was developed to improve research for lung inflammatory diseases. ... Human alveolar macrophages have been challenging to study because they reside deep in the lungs and are hard to access. The new ... "I am excited to see the full potential of the alveolar macrophage-like cells and if they can be integrated into next-generation ... The findings are published in mBio in an article titled, " A new tractable method for generating human alveolar macrophage-like ...
Macrophages were not the only immune cells affected; while the proliferation of T cells was unchanged, their antitumor activity ... It was demonstrated that cancer stem cells cause macrophages, which are immune cells in charge of the initial stage of cancer ... ice with healthy immune systems are exposed to cancer stem cells, which cause macrophage ageing and tumour growth. ... Our results indicate that drugs targeting senescent macrophages could be a treatment for cancer--an unprecedented development ...
Tumor-assisting macrophages: The M2 phenotype. TAMs can take on a variety of roles to support cancer cell survival and ... Tumor-killing macrophages: The M1 phenotype. TAMs have the potential to aid antitumor immune responses by presenting cancer ... Infographic: Tumor-Associated Macrophages and Cancer. The immune cells play various roles in the tumor-some that assist ... umors use chemokine signals to draw monocytes and tissue-resident macrophages into the tumor microenvironment, where the cells ...
In this review, we discuss the origin and development of macrophages and their role in the intestinal inflammatory response or ... In this review, we discuss the origin and development of macrophages and their role in the intestinal inflammatory response or ... In addition, the effects of macrophages in the occurrence and development of inflammatory bowel disease (IBD), and their role in ... A clear understanding of the function of macrophages, as well as their role in pathogens and inflammatory response, will ...
... Proc Assoc Am Physicians. 1998 Jul-Aug; ... Recent work with transgenic mice demonstrated that disruption of the production of granulocyte-macrophage colony-stimulating ... by dysfunction of the alveolar macrophage. Local production of GM-CSF corrects the alveolar proteinosis in the GM-CSF knockout ... by its actions on the clearance of surfactant lipids and proteins by the alveolar macrophage. These findings may have important ...
Macrophage Detachment Solution DXF 250 ml; find Sigma-Aldrich-C41330 MSDS, related peer-reviewed papers, technical documents, ... The Macrophage Detachment Solution DXF is non-enzymatic, defined and animal component-free. It was especially designed for the ... In Vitro Differentiation of Human PBMC Derived Monocytes into M1 or M2 Macrophages in a Serum-free and Xeno-free Cell Culture ... Generate phagocytic M1 and M2 macrophages from human PBMCs in 10 days in serum-free and xeno-free cell culture media. Explore ...
Iron content in human alveolar macrophages. JL Corhay, G Weber, T Bury, S Mariz, I Roelandts, MF Radermecker ... Iron content in human alveolar macrophages Message Subject (Your Name) has sent you a message from European Respiratory Society ... Both techniques have been used to measure iron in alveolar macrophages gathered by bronchoalveolar lavage. The purpose of this ... Using the PIXE method, we measured the iron content of alveolar macrophages in healthy subjects, with and without occupational ...
In the present study we demonstrate that chronic filarial infection modulates macrophages to a less pro-inflammatory phenotype ... play a crucial role in triggering the correct macrophage response via TLR2. Thus, our observations suggest that helminths and ...
These macrophages expressed prominent levels of monocarboxylate transporter-4 (MCT-4), which is specialized in the secretion of ... Relevance to MS was corroborated by the strong expression of MCT-4, EMMPRIN, and LDHA in perivascular macrophages in MS brains ... The functional relevance of MCT-4-EMMPRIN interaction was confirmed by lower macrophage transmigration in culture using the MCT ... These results detail the metabolism of macrophages for transmigration from perivascular cuffs into the CNS parenchyma and ...
Yet the tattoos appearance did not change when the macrophages were killed off. The team determined that the dead macrophages ... "We think that, when tattoo pigment-laden macrophages die during the course of adult life, neighboring macrophages recapture the ... The researchers found that dermal macrophages were the only cell type to take up pigment when they tattooed the mices tails. ... "Tattoo removal can be likely improved by combining laser surgery with the transient ablation of the macrophages present in the ...
Macrophage Infection by HIV: Implications for Pathogenesis and Cure, from October 13-14, 2021. This video is Day 1. ... Macrophages that may be close to blood vessels, macrophages that may be close to nerves, macrophages that may reside in ... So this meeting will cover macrophages as targets for HIV infection, macrophages as mediators of inflammation, macrophages as a ... We also isolate macrophages from one of our control wells, which only contain macrophages, to look at DNA and RNA from the cell ...
An amino-bisphosphonate targets MMP-9-expressing macrophages and angiogenesis to impair cervical carcinogenesis. ... An amino-bisphosphonate targets MMP-9-expressing macrophages and angiogenesis to impair cervical carcinogenesis. ... expressed by infiltrating macrophages, similar to what has been observed in humans. Preclinical trials sought to target MMP-9 ... ZA suppressed MMP-9 expression by infiltrating macrophages and inhibited metalloprotease activity, reducing association of VEGF ...
A new study investigates whether SARS-CoV-2 can enter and replicate in macrophages and whether ACE2 is involved in these ... A new Science Signaling study evaluates whether macrophage ACE2 expression regulates the susceptibility of macrophages against ... However, macrophages support the early phase of viral infection, particularly the entry and synthesis of new viral RNA and ... Study: Macrophage ACE2 is necessary for SARS-CoV-2 replication and subsequent cytokine responses that restrict continued virion ...
Human macrophages are produced by culturing primary peripheral blood monocytes in RPMI 1640 with FBS, M-CSF and IL-4 and are ... Start with ready-to-use, ethically sourced, primary human macrophages. With personalized service, custom products, flexible ... Mechanistically, TMEM173 binding to ITPR1 controls calcium release from the endoplasmic reticulum in macrophages and monocytes ... and IL-4 for 5 days to generate macrophages. IL-4 in combination with M-CSF may induce M2a-like properties. Cells are collected ...
To investigate whether Trib3 regulates the polarisation state of macrophages, bone marrow derived macrophages (BMDMs) were ... However, adipocyte and macrophage numbers were increased in the Trib3KO mice. No significant differences in gene expression of ... Adipose tissue macrophages (CD11b+ATMs) were isolated from adipose tissue using magnetics beads and the expression of ... Conclusion The results obtained in this pilot study suggest a role for Trib3 in macrophages due to expression differences ...
Allan Mowat on Intestinal macrophages - heterogeneity, origins and functions, part of a collection of multimedia lectures. ... Intestinal macrophages - heterogeneity, origins and functions. *Prof. Allan Mowat - University of Glasgow, UK ... Distinct populations of macrophages control each of these functions. *Novel strategies for characterizing mouse intestinal ... Macrophages play essential roles in both homeostasis and inflammation in the intestine ...
Identification of interferon stimulated genes that control Toxoplasma in pig macrophages. Objective ... The fundamental insights that we will obtain will provide a deeper understanding ISG-Toxoplasma interactions in pig macrophages ... Identification of interferon stimulated genes that control Toxoplasma in pig macrophages ... expressed ISGs directly control Toxoplasma in pig macrophages using pig pluripotent stem cell-derived macrophages and high ...
Fate-Mapping Macrophages: From Ontogeny to Functions.. Ahlback A, Gentek R. Fate-Mapping Macrophages: From Ontogeny to ...
  • Coculture of macrophages with regulatory T cells (Tregs) caused differentiation of macrophages toward Mreg phenotype. (wikipedia.org)
  • However, at least two distinct macrophage populations have been identified: the classically activated M1 phenotype and the alternative activated M2 phenotype [ 6 ]. (hindawi.com)
  • In vivo , the identification of macrophage phenotype is complicated since macrophages are obviously exposed to a microenvironment that is more complex respect to cell culture conditions, and they display characteristics that do not conform to the in vitro defined phenotypic categories. (hindawi.com)
  • Therefore, in the last years characterization of macrophage phenotype in vivo during tissue repair has been a matter of active investigation. (hindawi.com)
  • We developed cell culture conditions that mimic the lung alveolar environment in humans using lung lipids, that is, Infasurf (calfactant, natural bovine surfactant) and lung-associated cytokines (granulocyte macrophage colony-stimulating factor, transforming growth factor-β, and interleukin 10) that facilitate the conversion of blood-obtained monocytes to an AM-like (AML) phenotype and function in tissue culture. (genengnews.com)
  • No significant differences in gene expression of classical polarisation markers were found in ATMs, however differential gene expression of proteins involved in macrophage function and phenotype was found in Trib3KO BMDMs compared to wild type results among the different polarisation states. (bmj.com)
  • Conclusion The results obtained in this pilot study suggest a role for Trib3 in macrophages due to expression differences measured in Trib3 deficient BMDMs, linked to a pro-inflammatory macrophage phenotype. (bmj.com)
  • Cardiac macrophages are heterogenous in phenotype and functions, which has been associated with differences in their ontogeny. (elifesciences.org)
  • Alternative polarization with interleukin-4 and IL-13 led to a macrophage phenotype characterized by increased tissue factor (TF) production and release and by an increase in extracellular vesicle production. (haematologica.org)
  • In conclusion, we provide evidence that proinflammatory polarization of macrophages does not lead to enhanced procoagulatory function, whereas alternative polarization of macrophages leads to an increased expression of TF and increased production of TF bearing extracellular vesicles by these cells suggesting a procoagulatory phenotype of alternatively polarized macrophages. (haematologica.org)
  • This macrophage polarization can be simulated in vitro using lipopolysaccharide (LPS) and interferon (IFN)-γ stimulation for a proinflammatory subset termed classical activation and stimulation with interleukin (IL)-4 and IL-13 for an alternative polarization phenotype. (haematologica.org)
  • 4 Upon polarization, macrophages react to the respective stimulus with the expression of a distinct phenotype. (haematologica.org)
  • The primary aim of the present study was to determine the immunological phenotype of NSCLC-associated macrophages. (ersjournals.com)
  • CD68 + macrophages expressing markers of a cytotoxic M1 phenotype or a noncytotoxic M2 phenotype were identified in the islets and stroma of surgically resected tumours from 20 patients with extended survival (median 92.7 months) and 20 with poor survival (median 7.7 months), using immunohistochemistry. (ersjournals.com)
  • Macrophages infiltrating the tumour islets in nonsmall cell lung cancer were predominantly of the M1 phenotype in patients with extended survival. (ersjournals.com)
  • The M1 phenotype (classically activated) macrophages are thought to be induced by interferon-γ, with or without lipopolysaccharide and tumour necrosis factor (TNF)-α, and exert a cytotoxic effect against cancer cells. (ersjournals.com)
  • Additionally, M2 macrophages have been traditionally thought of as being the predominant macrophage phenotype in solid tumours (tumour-associated macrophages). (ersjournals.com)
  • Macrophage activation has been described as a dynamic process: the same cell may initially induce proinflammatory and cytotoxic reactions and later may take part in the resolution of inflammation and wound healing [ 9 ]. (hindawi.com)
  • A fundamental problem in understanding how macrophages regulate inflammation is their phenotypical and functional heterogeneity. (nature.com)
  • Solinas G, Germano G, Mantovani A, Allavena P. Tumor-associated macrophages (TAM) as major players of the cancer-related inflammation. (springer.com)
  • In addition, the effects of macrophages in the occurrence and development of inflammatory bowel disease (IBD), and their role in inducing fibrosis, activating T cells, reducing colitis, and treating intestinal inflammation were also reviewed in this paper. (frontiersin.org)
  • Macrophages are phagocytes found in tissues and maintain tissue homeostasis, regulate inflammation, and play a significant role in host protection. (frontiersin.org)
  • The meeting will also highlight recent work on macrophage inflammation in the context of SARS-CoV-2 infection and work from recently-funded NIMH, NIDA, and NINDS investigators. (nih.gov)
  • A panel will discuss research gaps and priorities related to human macrophage reservoirs and inflammation/comorbidities and treatment strategies for HIV remission and cure. (nih.gov)
  • The metabolic and immune responses to obesity affect many organs and particularly the adipose tissue where macrophages are key effector cells modulating inflammation through the signals received from adipocytes. (bmj.com)
  • 2 Upon inflammation, the pool of resident macrophages gets quickly replaced by macrophages derived from circulating monocytes. (haematologica.org)
  • Researchers at Texas Biomedical Research Institute report they have created a new cell culture model for human alveolar macrophages in the lab. (genengnews.com)
  • The findings are published in mBio in an article titled, " A new tractable method for generating human alveolar macrophage-like cells in vitro to study lung inflammatory processes and diseases . (genengnews.com)
  • Alveolar macrophages (AMs) are unique lung resident cells that contact airborne pathogens and environmental particulates," wrote the researchers. (genengnews.com)
  • Human alveolar macrophages have been challenging to study because they reside deep in the lungs and are hard to access. (genengnews.com)
  • The generated cells are 94% genetically similar to human alveolar macrophages collected from lung washes. (genengnews.com)
  • I am excited to see the full potential of the alveolar macrophage-like cells and if they can be integrated into next-generation lung organoids," Schlesinger added. (genengnews.com)
  • Recent work with transgenic mice demonstrated that disruption of the production of granulocyte-macrophage colony-stimulating factor (GM-CSF) or the common beta-subunit of the GM-CSF receptor caused alveolar proteinosis that was histologically similar to that seen in human patients. (nih.gov)
  • The defect in surfactant homeostasis is caused by decreased surfactant clearance, mediated (at least in part) by dysfunction of the alveolar macrophage. (nih.gov)
  • These studies demonstrate the previously unanticipated role of GM-CSF signaling in surfactant homeostasis, mediated (at least in part) by its actions on the clearance of surfactant lipids and proteins by the alveolar macrophage. (nih.gov)
  • Both techniques have been used to measure iron in alveolar macrophages gathered by bronchoalveolar lavage. (ersjournals.com)
  • Using the PIXE method, we measured the iron content of alveolar macrophages in healthy subjects, with and without occupational exposure to iron dust, and in patients with pulmonary diseases (chronic obstructive pulmonary disease (COPD), lung cancer, Goodpasture's syndrome). (ersjournals.com)
  • Mean iron content of blood monocytes, lymphocytes and neutrophils of eight healthy subjects was significantly lower than that of alveolar macrophages. (ersjournals.com)
  • cSiO2 causes phagolysosomal membrane permeabilization (LMP) and NLRP3 inflammasome activation in alveolar macrophages. (nih.gov)
  • Microglia are brain-resident macrophages. (frontiersin.org)
  • They are often studied independently from other tissue-resident macrophages, probably because they are unique in some aspects, most notably in their isolation from the rest of the body through the blood brain barrier (BBB). (frontiersin.org)
  • But how different are microglia really from other tissue resident macrophages in terms of origin, lineage, and identity? (frontiersin.org)
  • In the next sections, we will review evidence about the origin, lineage, identity, and population dynamics of microglia compared to other tissue-resident macrophages and highlight commonalities and differences. (frontiersin.org)
  • proposed the "mononuclear phagocyte system" theory, by which tissue-resident macrophages were assumed to derive from blood-circulating monocytes and to differentiate within the host tissue ( 1 ). (frontiersin.org)
  • umors use chemokine signals to draw monocytes and tissue-resident macrophages into the tumor microenvironment, where the cells become tumor-associated macrophages (TAMs). (the-scientist.com)
  • Originating either from monocytes that come from bone marrow, or tissue-resident macrophages that arise during embryonic development, they can repress antitumor immunity by secreting cytokines such as IL-10, which blocks dendritic cell activation, and TGF-β, which blunts T-cell responses (1). (the-scientist.com)
  • Recently, we collected evidence that survival of enteric neurons is guaranteed by a unique subpopulation of resident macrophages closely associated to the ENS and expressing a typical neuroprotective / -supportive transcriptome. (europa.eu)
  • In summary, resident macrophages orchestrate inflammatory responses improving cardiac remodelling, while recruited macrophages determine infarct size after I/R injury. (elifesciences.org)
  • Using state-of-the-art fate-mapping models and genetic and pharmacological targeting approaches, the authors provide solid evidence that the absence of resident macrophages do not influence infarct size but instead alter the immune cell crosstalk in response to injury. (elifesciences.org)
  • However, the functional evaluation of resident macrophages is limited by potential off target effects in ∆FIRE mice. (elifesciences.org)
  • For example, microglia , the longest known tissue -resident macrophages , show a highly branched process tree with which they continuously scan their environment . (bvsalud.org)
  • Computational image analysis allows to quantify morphology and/or motility from images of tissue -resident macrophages . (bvsalud.org)
  • Axolotl WIKIMEDIA, STAN SHEBS Macrophages, a type of immune cell that travels throughout tissues gobbling up bacteria and dead cells, plays a pivotal role in limb regeneration in a salamander called the axolotl, according to new research published this week (May 20) in The Proceedings of the National Academy of Sciences . (the-scientist.com)
  • The presence of immune cells, such as macrophages, in metabolic tissues suggests dynamic, ongoing crosstalk between these two regulatory systems. (nih.gov)
  • Beyond their role in innate immunity, macrophages play increasingly defined roles in orchestrating the healing of various injured tissues. (hindawi.com)
  • Macrophages are essential for the efficient healing of numerous tissues. (hindawi.com)
  • We here try to address these questions by collecting answers from microglia's cousins, the macrophages that reside in other tissues. (frontiersin.org)
  • Macrophage heterogeneity in tissues: phenotypic diversity and functions. (springer.com)
  • A team of researchers led by Sandrine Henri and Bernard Malissen of the Centre d'Immunologie de Marseille-Luminy developed a genetically engineered mouse that allowed them to kill the macrophages that reside in the dermis and certain other tissues. (newswise.com)
  • Macrophages exist in nearly all tissues and are produced when white blood cells called monocytes leave the blood and differentiate in a tissue-specific manner. (zmescience.com)
  • Macrophages reside in all tissues of the body and each population of macrophages within a tissue can take on specialized functions that are tuned to the developmental and functional requirements of that tissue. (haematologica.org)
  • Monocytes become macrophages when they move from the bloodstream to the tissues. (msdmanuals.com)
  • Macrophages stay in the tissues. (msdmanuals.com)
  • Through mechanisms that have not yet been well elucidated, this impairment in cytotoxic function leads to an excessive expansion and activation of cytotoxic cells, with hypersecretion of proinflammatory cytokines such as interferon (IFN)-γ, tumor necrosis factor (TNF)-α, interleukin (IL)-6, IL-10, and macrophage-colony-stimulating factor (M-CSF). (medscape.com)
  • Macrophages participate in all the different phases of tissue repair: they can promote phagocytosis of cellular debris and apoptotic neutrophils and produce cytokines that may help orchestrate the healing response. (hindawi.com)
  • However, due to the release of proinflammatory cytokines and cytotoxic radical species, uncontrolled activity of macrophages may also be detrimental to tissue repair. (hindawi.com)
  • Macrophages play a significant role in many processes, such as the human immune function, parasite infection, and tissue remodeling by secreting cytokines and producing reactive oxygen and nitrogen intermediates. (frontiersin.org)
  • During severe infection, macrophages have been identified to be the key source of proinflammatory cytokines. (news-medical.net)
  • In macrophage-colony stimulating factor (MCSF) generated macrophages, Sia + IgG induced increased production of numerous cytokines/ chemokines including IL-6, TNFα, CXCL1, and IL-10, and the expression of the macrophage surface marker CD163. (scirp.org)
  • D. Bruder, E. , O. Richards, J. , M. Michel, K. and Oaks, M. (2016) Differential Effects of Alternative Glycoforms of IgG on Human Monocytes and Macrophages: Sialylated IgG Induces Novel Expression Signatures of Cell Surface Markers, Cytokines, and Chemokines. (scirp.org)
  • Classically activated M1 macrophages are induced in vitro by IFN γ , alone or in concert with microbial stimuli (e.g. (hindawi.com)
  • Importantly, stimulation of T cells by Cd44v7- deficient macrophages induced upregulation of Foxp3 in vitro , while cotransfer of Cd44 WT macrophages into Cd44v7- deficient mice reduced Foxp3 + T-cell counts and caused colitis. (nature.com)
  • Since human monocyte-derived macrophages (HMDMs) do not express ACE2, these cells were used as in vitro models for ACE2-negative macrophages. (news-medical.net)
  • Consistent with previous in vitro studies, the current study revealed that SARS-CoV-2 did not replicate in BAL macrophages or HMDM. (news-medical.net)
  • To investigate whether Trib3 regulates the polarisation state of macrophages, bone marrow derived macrophages (BMDMs) were isolated and polarised in vitro to further characterise their pro- or anti- inflammatory properties. (bmj.com)
  • In vitro systems are used to differentiate these cells into diverse macrophage subsets to mimic the populations observed in vivo . (biorxiv.org)
  • Regulatory macrophages (Mregs) represent a subset of anti-inflammatory macrophages. (wikipedia.org)
  • Indeed, several human diseases are characterized by attenuated repair responses and imbalances in the inflammatory response with increased number of infiltrating macrophages [ 3 - 5 ]. (hindawi.com)
  • single-cell sequencing combined with new macrophage-specific genetic tools has provided unexpected insights into the tissue-specific and inflammatory diversity of macrophages. (nature.com)
  • In addition, a new mechanism of inflammatory control of macrophage diversity based on lactate-mediated histone modifications was discovered. (nature.com)
  • An unexpected 'timing' mechanism mediated by lactate seems to control macrophage inflammatory transitions. (nature.com)
  • Pro-inflammatory M1 macrophages hold the potential to engulf tumor cells. (springer.com)
  • In contrast, anti-inflammatory M2 macrophages, which are predominantly present in tumors, potentiate tumor progression and immune escape. (springer.com)
  • Macrophages, which are functional plasticity cells, have the ability to phagocytize and digest foreign substances and acquire pro-(M1-like) or anti-inflammatory (M2-like) phenotypes according to their microenvironment. (frontiersin.org)
  • A clear understanding of the function of macrophages, as well as their role in pathogens and inflammatory response, will delineate the next steps in the treatment of intestinal inflammatory diseases. (frontiersin.org)
  • In this review, we discuss the origin and development of macrophages and their role in the intestinal inflammatory response or infection. (frontiersin.org)
  • Mycobacteria are opportunistic microbes that reside and multiply in host macrophages, mainly by inhibition of host pro-inflammatory response. (europa.eu)
  • The lysosome's role in NLRP3 inflammasome activity and its contributions to macrophage homeostasis make it of significant interest in the development of therapeutic targets for chronic inflammatory human health conditions. (nih.gov)
  • Besides scavenging debris, promoting angiogenesis, tissue remodeling and repair, alternatively activated macrophages are able to fine tune inflammatory responses. (haematologica.org)
  • Pulmonary cytotoxicity, inflammatory cell influx, and relative mRNA expression of genes related to macrophage function and recruitment (Ccl2, Ccl22, and Arg1) were increased as a result of MWCNT exposure but not affected by OPN deficiency. (cdc.gov)
  • Whether the sequential presence of two different macrophage populations results from a dynamic shift in macrophage polarization or from the recruitment of new circulating monocytes is a subject of ongoing debate. (hindawi.com)
  • Major differences were reported in gene expression between human and mouse macrophages in the setting of lung cancer, whereas gene expression in other immune cell populations closely correlated between species. (nature.com)
  • How can we determine (and potentially target) a pathogenic or protective macrophage function if there are multiple types of macrophage in any given tissue site and if these populations temporally change their phenotypes in response to local extrinsic cues? (nature.com)
  • In the first part of this review, we will discuss similarities and differences in the identity of microglia and other macrophages, taking into account their developmental origin and the maintenance of the adult populations. (frontiersin.org)
  • However, the mechanistic differences when SARS-CoV-2 infects other cell types, such as macrophage populations, are poorly understood. (news-medical.net)
  • Once established, both the peritubular and interstitial macrophage populations exhibit a long life span and a low turnover in the steady state," the authors wrote. (zmescience.com)
  • In contrast, continuous CSF1R inhibition led to depletion of both resident and recruited macrophage populations. (elifesciences.org)
  • These findings attribute distinct beneficial effects to different macrophage populations in the context of myocardial infarction. (elifesciences.org)
  • Macrophages are associated with innate immunity that protects the respiratory tracts during viral infection. (news-medical.net)
  • Macrophages are important effectors of innate immunity. (elifesciences.org)
  • Macrophages are cells of the innate immune system which play numerous and vastly different functions within the body. (haematologica.org)
  • Here, we report that bone marrow derived macrophages and human monocyte-derived macrophages differentiated with either M-CSF or GM-CSF show different levels of LRRK2 activation after membrane damage. (biorxiv.org)
  • Here, we discuss how changes in the recruitment and activation of macrophages contribute to metabolic homeostasis. (nih.gov)
  • Heterogeneity and plasticity have been demonstrated to be hallmarks of macrophages. (hindawi.com)
  • Heterogeneity and plasticity of macrophages could explain these apparently contrasting roles in tissue healing. (hindawi.com)
  • However, the twin problems of dissecting macrophage phenotypical heterogeneity and specific function at a given time and place remain. (nature.com)
  • Fortunately, technological innovations are finally allowing us to address these issues, and in 2019 single-cell approaches and new fate mapping tools greatly advanced our understanding of macrophage heterogeneity. (nature.com)
  • Intestinal macrophages - heterogeneity, origins and functions [Video file]. (hstalks.com)
  • Monocyte and macrophage plasticity in tissue repair and regeneration. (springer.com)
  • The team determined that the dead macrophages release the pigment into their surroundings, where, over the following weeks, it is taken up by new, monocyte-derived macrophages before it can disperse. (newswise.com)
  • Genomic deletion of a fms-intronic regulatory element (FIRE) in the Csf1r locus resulted in specific absence of resident homeostatic and antigen-presenting macrophages, without affecting the recruitment of monocyte-derived macrophages to the infarcted heart. (elifesciences.org)
  • Specific absence of homeostatic, monocyte-independent macrophages altered the immune cell crosstalk in response to injury and induced proinflammatory neutrophil polarization, resulting in impaired cardiac remodelling without influencing infarct size. (elifesciences.org)
  • To understand if polarization of macrophages can lead to a procoagulant macrophage subset we polarized human monocyte derived macrophages to a proinflammatory and an alternative activation state. (haematologica.org)
  • The present study examined the effects of Sia + IgG on human peripheral blood mononuclear cell (PBMC)-derived monocyte and macrophage surface marker expression and cytokine/chemokine secretion. (scirp.org)
  • M-CSF and GM-CSF differentiated human blood monocytes are often used to generate monocyte-derived macrophages as a model for tissue macrophages. (biorxiv.org)
  • [ 1 , 2 ] Macrophage activation syndrome is characterized by pancytopenia, liver insufficiency, coagulopathy, and neurologic symptoms and is thought to be caused by the activation and uncontrolled proliferation of T lymphocytes and well-differentiated macrophages, leading to widespread hemophagocytosis and cytokine overproduction. (medscape.com)
  • The initial tissue reaction to infection is a focal, intensely suppurative necrosis consisting largely of accumulations of polymorphonuclear leukocytes, followed by invasion of macrophages, epithelioid cells, and lymphocytes. (cdc.gov)
  • The analyses implicated cellular and molecular targets of ZA's actions: ZA suppressed MMP-9 expression by infiltrating macrophages and inhibited metalloprotease activity, reducing association of VEGF with its receptor on angiogenic endothelial cells. (jci.org)
  • Given its track record in clinical use with limited toxicity, ZA holds promise as an "unconventional" MMP-9 inhibitor for antiangiogenic therapy of cervical cancer and potentially for additional cancers and other diseases where MMP-9 expression by infiltrating macrophages is evident. (jci.org)
  • It was demonstrated that cancer stem cells cause macrophages, which are immune cells in charge of the initial stage of cancer cell death, to undergo senescence. (newkerala.com)
  • More recently, however, researchers have suggested that macrophages-specialized immune cells that reside in the dermis-are attracted to the wound inflicted by the tattoo needle and gobble up the tattoo pigment just as they would normally engulf an invading pathogen or piece of a dying cell. (newswise.com)
  • The Parkinson's Disease (PD) kinase LRRK2 is highly expressed in immune cells such as macrophages. (biorxiv.org)
  • In general, macrophages are a very dynamic and plastic cell type and can be divided into two main groups: classically activated macrophages (M1) and alternatively activated macrophages (M2). (wikipedia.org)
  • Surprisingly, Mregs resemble classically activated macrophages more than alternatively activated macrophages, due to higher biochemical similarity. (wikipedia.org)
  • Unlike classically activated macrophages, Mregs produce low levels of IL-12, which is important because IL-12 induces differentiation of naïve helper T cells to Th1 cells which produce high levels of IFNγ. (wikipedia.org)
  • In particular, we focus our discussion on the pathogenic and protective functions of classically and alternatively activated macrophages, respectively, in experimental models of obesity and metabolic disease. (nih.gov)
  • In this review, we intend to summarize the current knowledge on the many functions of autophagy proteins in cell defenses with a focus on bacteria-macrophage interaction. (frontiersin.org)
  • Unlike enzyme-based solutions, the Macrophage Detachment Solution DXF does not alter cell surface proteins and neutralization is not necessary.Note: The Macrophage Detachment Solution DXF is not suitable for the dissociation of other adherent cell types. (sigmaaldrich.com)
  • The large number of macrophages in the intestinal tract, play a significant role in maintaining the homeostasis of microorganisms on the surface of the intestinal mucosa and in the continuous renewal of intestinal epithelial cells. (frontiersin.org)
  • We here investigated macrophage lineages and ablated tissue macrophages in homeostasis and after I/R injury in a CSF1R-dependent manner. (elifesciences.org)
  • Stein M, Keshav S, Harris N, Gordon S. Interleukin 4 potently enhances murine macrophage mannose receptor activity: a marker of alternative immunologic macrophage activation. (springer.com)
  • Several molecules expressed by macrophages and T cells including interleukin-12 (IL-12), interferon-γ, IL-10, and Foxp3 are critical for the intestinal balance between immune attack and tolerance. (nature.com)
  • M1 macrophages are associated with the expression of interleukin (IL)-1, -12, TNF-α and inducible nitric oxide synthase (iNOS) 7 . (ersjournals.com)
  • The first signal promotes the differentiation of monocytes to macrophages and the second signal promotes immunosuppressive functions. (wikipedia.org)
  • while the proliferation of T cells was unchanged, their antitumor activity was suppressed due to the immunosuppressive factors produced by senescent macrophages. (newkerala.com)
  • The team also demonstrated that supplementing the mice inoculated with cancer stem cells with a molecule called nicotinamide mononucleotide resulted in the proliferation of non-senescent macrophages and reduced the immunosuppressive factors produced by senescent macrophages, preventing tumour growth and leading to increased survival times in mice. (newkerala.com)
  • The modular nature of our platform offers multiple therapeutic possibilities, and our collaboration with Macrophage Therapeutics may enable the development of Accurins that target activated macrophages, which in cancer, help create an immunosuppressive microenvironment. (businesswire.com)
  • Disease-associated macrophages generally play a pro-tumoral role and are immunosuppressive, preventing the immune system from mounting an attack on tumor cells. (businesswire.com)
  • We propose that the negative prognostic value related to CD169 + macrophages in PTs is a consequence of an immunosuppressive tumor environment rich in TLSs, T regs and B regs . (lu.se)
  • We provide a novel treatment strategy against pediatric brain tumors as well as increased understanding of how to break complex immunosuppressive macrophage derived mechanisms. (lu.se)
  • M2a, mainly known as alternatively activated macrophages, are macrophages associated with tissue healing due to the production of components of extracellular matrix. (wikipedia.org)
  • Out of all macrophages, Mregs show the highest expression of MHC II molecules and co-stimulatory molecules (CD80/CD86), which differentiates them from the alternatively activated macrophages, which show a very low expression of these molecules. (wikipedia.org)
  • Mregs also differ from alternatively activated macrophages by producing high levels of nitric oxide and low arginase activity. (wikipedia.org)
  • Lastly, they differ in the expression of FIIZ1 (Resistin-like molecule alpha1) and YM1 which are differentiation markers present on alternatively activated macrophages. (wikipedia.org)
  • Alternatively activated M2 macrophages comprise cells exposed to low concentrations of M-CSF in the presence of IL-4, IL-13, or IL-10. (hindawi.com)
  • In addition, also TF activity was enhanced in extracellular vesicles of alternatively polarized macrophages. (haematologica.org)
  • 7 In contrast, alternatively activated macrophages are characterized by increased expression of IL-10 and of scavenger receptors. (haematologica.org)
  • M2 (alternatively activated) macrophages are thought to be modulated by IL-4 and -13 and associated with tumour formation. (ersjournals.com)
  • Herein, we introduce the role of macrophages in tumor progression, summarize the recent advances in macrophage-centered anticancer therapy, and discuss their challenges as well as future applications. (springer.com)
  • Here, we review the many functions of autophagy in bacterial infections with a focus on macrophages, the first line of host defenses, and the replicative niche of numerous pathogens. (frontiersin.org)
  • So on day one, Session 1 will focus on macrophages and the immune system during HIV and SARS-CoV-2 infection, Session 2 will focus on challenges to study HIV/SIV reservoirs, and Session 3 will really have the highlights from recent awardees of the NIMH/NINDS/NIDA Myeloid Reservoir RFA. (nih.gov)
  • Our data extended prior observations of Sia + IgG on DC function and showed that Sia + IgG was able to differentially modulate multiple pathways in monocytes and macrophages. (scirp.org)
  • Roles of autophagy machinery in macrophage antibacterial defenses. (frontiersin.org)
  • It has also been described that one of the key roles of M2 macrophages is to promote angiogenesis 7 , 10 . (ersjournals.com)
  • In this paper, we discuss the current available information about the role that different phenotypes of macrophages plays after injury and during the remodelling phase in different tissue types, with particular attention to the skeletal muscle. (hindawi.com)
  • and second, macrophages are sufficiently plastic that they can adopt tissue-specific phenotypes independent of their origin. (nature.com)
  • The role of macrophage phenotypes in tumour progression has been extensively reviewed 6 - 10 . (ersjournals.com)
  • Recent evidence, however, supports a dual role for macrophages in the regulation of tumour proliferation and immune control 10 , 16 , and indicates that the local tissue microenvironment plays a critical role in determining cell phenotypes. (ersjournals.com)
  • Targeting macrophages to modulate the tumor immune microenvironment can ameliorate the tumor-associated immunosuppression and elicit an anti-tumor immune response. (springer.com)
  • Based on the expression of CD206 mannose receptors on disease-associated macrophages, BIND and Macrophage plan to conduct joint research to develop a CD206 targeted Accurin nanoparticle that is capable of concentrating various therapeutic payloads to the tumor microenvironment. (businesswire.com)
  • By coupling Accurins with Macrophage's well-credentialed CD206 targeting ligand, we may be able to treat macrophage-mediated diseases through increased uptake, and concentration, of targeted therapeutic payloads in the tumor microenvironment. (businesswire.com)
  • Here, macrophages and T cells accumulate in the lamina propria of the intestinal tract, activate each other, destroy the regular architecture of the gut wall, and disturb the intestinal function. (nature.com)
  • The lamina propria of the small intestine is the main site of the intestinal immune system, which contains a large number of macrophages, CD4 T cells, and dendritic cells. (frontiersin.org)
  • A single-cell atlas of mouse brain macrophages reveals unique transcriptional identities shaped by ontogeny and tissue environment. (nature.com)
  • Macrophages are widespread and reside in many different organs, where they fulfill different functions. (frontiersin.org)
  • In contrast, CD44v7 absence on macrophages in recipient mice prevented colitis. (nature.com)
  • However, adipocyte and macrophage numbers were increased in the Trib3KO mice. (bmj.com)
  • Studying mice, the French researchers led by Michael Sieweke found peritubular macrophages only appear two weeks after the mice were born, which coincides with the rodent's puberty. (zmescience.com)
  • We have shown that MB and other human brain tumors express high levels of CD24, and intratumoral delivery of AF16 cured mice with gliomas, possibly via macrophage modulation. (lu.se)
  • Macrophages, as one of the most abundant tumor-infiltrating cells, play an important role in tumor development and metastasis. (springer.com)
  • The frequency and polarization of tumor-associated macrophages (TAMs) correlate with disease progression, tumor metastasis, and resistance to various treatments. (springer.com)
  • Besides, biomimetic carriers based on macrophages have been extensively explored to prolong circulation, enhance tumor-targeted delivery, and reduce the immunogenicity of therapeutics to augment therapeutic efficacy. (springer.com)
  • Moreover, the genetic engineering of macrophages with chimeric antigen receptor (CAR) allows them to recognize tumor antigens and perform tumor cell-specific phagocytosis. (springer.com)
  • Komohara Y, Fujiwara Y, Ohnishi K, Takeya M. Tumor-associated macrophages: potential therapeutic targets for anti-cancer therapy. (springer.com)
  • Zhou XF, Liu XR, Huang L. Macrophage-mediated tumor cell phagocytosis: opportunity for nanomedicine intervention. (springer.com)
  • Tumor-associated macrophages: potential therapeutic strategies and future prospects in cancer. (springer.com)
  • Yang QY, Guo NN, Zhou Y, Chen JJ, Wei QC, Han M. The role of tumor-associated macrophages (TAMs) in tumor progression and relevant advance in targeted therapy. (springer.com)
  • At the same time, Tie2 + macrophages come together with cancer cells and blood vessel endothelial cells to form complexes, called tumor. (the-scientist.com)
  • On the other hand, phagosomes are formed when the pathogen is engulfed by macrophages and fuse with lysosomes to release enzymes and toxic substances, resulting in killing or having cytotoxic effects on bacteria and tumor cells. (frontiersin.org)
  • We here investigated the prognostic impact of tumor-infiltrating CD169 + macrophages in primary tumors (PTs) and their spatial relation to tumor-infiltrating B and T cells. (lu.se)
  • Brain tumors contain huge amounts of tumor-promoting macrophages, but these can be drug-modulated to kill tumor cells. (lu.se)
  • Perturbations of macrophage function and/or activation may result in impaired regeneration and fibrosis deposition as described in several chronic pathological diseases. (hindawi.com)
  • However, we have now extended these studies to use retroviral gene transfer methods to stably transduce macrophage precursors with these DNA constructs. (yourdictionary.com)
  • The other kind of macrophage is peritubular, named so because we can find it on the surface of seminiferous tubules that house sperm cell precursors. (zmescience.com)
  • 1 Some tissue macrophages and precursors are already established embryonically in the yolk sac and fetal liver before the onset of definitive hematopoiesis. (haematologica.org)
  • Lung single-cell signaling interaction map reveals basophil role in macrophage imprinting. (nature.com)
  • Based on the findings of single-cell RNA sequencing (scRNA-seq), viral RNA has been detected within macrophages in lung tissue and bronchoalveolar lavage (BAL) fluid from COVID-19 patients. (news-medical.net)
  • There is a marked survival advantage for patients with nonsmall cell lung cancer (NSCLC) expressing high numbers of macrophages in their tumour islets. (ersjournals.com)
  • Using two breast cancer patient cohorts, we show that CD169 + macrophages were spatially associated with the presence of B and T cell tertiary lymphoid-like structures (TLLSs) in both PTs and lymph node metastases (LNMs). (lu.se)
  • macrophages were spatially associated with the presence of B and T cell tertiary lymphoid-like structures (TLLSs) in both PTs and lymph node metastases (LNMs). (lu.se)
  • A new Science Signaling study evaluates whether macrophage ACE2 expression regulates the susceptibility of macrophages against SARS-CoV-2 entry and replication. (news-medical.net)
  • Macrophages are one of the first barriers of host defence against pathogens. (hindawi.com)
  • Macrophages are central to host defense against microbes, but intracellular pathogens have evolved to evade their antimicrobial functions. (harvard.edu)
  • A macrophage is a large white blood cell which gobbles foreign pathogens like bacteria. (zmescience.com)
  • A close review of the literature shows that microglia are not as coarsely distinct to other macrophages as one may think, yet there are some fine differences in how they behave in their local environment. (frontiersin.org)
  • This study provides important findings on the distinct functions of resident and recruited macrophages during cardiac healing after myocardial ischemia. (elifesciences.org)
  • Tumors protect themselves from macrophage-mediated killing by CD24 expression. (lu.se)
  • We propose that Trib3 may modulate macrophage polarisation and therefore modulate the immune response to promote obesity. (bmj.com)
  • We show that embryonic progenitors give rise to the interstitial macrophage population, whereas peritubular macrophages are exclusively seeded postnatally in the prepuberty period from bone marrow (BM)-derived progenitors. (zmescience.com)
  • Macrophage activation syndrome (MAS) is a life-threatening complication of rheumatic disease that, for unknown reasons, occurs much more frequently in individuals with systemic juvenile idiopathic arthritis (SJIA) and in those with adult-onset Still disease. (medscape.com)
  • Typically, patients with macrophage activation syndrome become acutely ill with the sudden onset of nonremitting high fever, profound depression in all 3 blood cell lines (ie, leukopenia, anemia, and thrombocytopenia), hepatosplenomegaly, lymphadenopathy, and elevated serum liver enzyme levels. (medscape.com)
  • The treatment of macrophage activation syndrome is traditionally based on the parenteral administration of high doses of corticosteroids. (medscape.com)
  • Macrophage activation syndrome is characterized by a highly stimulated but ineffective immune response. (medscape.com)
  • Recent studies have shown that MUNC 13-4 polymorphisms are associated with macrophage activation syndrome in some patients with SJIA. (medscape.com)
  • Macrophage activation syndrome--what's in a name! (jrheum.org)
  • Similar effect provoked interaction of macrophages and B1 B cells. (wikipedia.org)
  • Because the bacteria live within macrophage cells, treatment may be difficult, and the treatment periods may vary. (yourdictionary.com)
  • The cells were collected after the daughter was given granulocyte macrophage colony stimulating factor for 5 days to boost her white blood cells. (yourdictionary.com)
  • The earliest detectable lesions, called fatty streaks, contain macrophage foam cells that are derived from recruited monocytes. (yourdictionary.com)
  • Then, we will discuss several principles that govern macrophage phagocytosis of apoptotic cells (efferocytosis), including the existence of redundant recognition mechanisms ("find-me" and "eat-me") that lead to a tight coupling between apoptosis and phagocytosis. (frontiersin.org)
  • Because it is such a diverse population of cells, a fundamental question is whether they have a common precursor or whether each macrophage population develops from a different precursor. (frontiersin.org)
  • Macrophages are also experts at phagocytosing and degrading foreign cells, including cancer cells (2). (the-scientist.com)
  • ice with healthy immune systems are exposed to cancer stem cells, which cause macrophage ageing and tumour growth. (newkerala.com)
  • further investigation showed that macrophages cultured with cancer stem cells exhibit senescence or cellular ageing. (newkerala.com)
  • These macrophages expressed prominent levels of monocarboxylate transporter-4 (MCT-4), which is specialized in the secretion of lactate from glycolytic cells. (jci.org)
  • Over the following few weeks, these cells are replaced by new macrophages derived from precursor cells known as monocytes. (newswise.com)
  • Although CD4 T cells are the main targets of HIV infection, macrophages also become infected and resist the cytopathic effects of infection, contributing potentially to HIV reservoir persistence. (nih.gov)
  • In contrast to epithelial cells, human macrophages manifest an abortive replication cycle for many viruses, such as rhinovirus and seasonal influenza A viruses. (news-medical.net)
  • Thus, macrophages not only inhibit viral dissemination but can sense infected cells and infectious viral particles to subsequently elicit antiviral and proinflammatory programs. (news-medical.net)
  • Guided by these and other preliminary data, this project will address the gap in our current knowledge of how ISGs control Toxoplasma in pig cells by systematically identifying which and how the differentially expressed ISGs directly control Toxoplasma in pig macrophages using pig pluripotent stem cell-derived macrophages and high throughput ISG functional screening tools that we have recently developed. (usda.gov)
  • Two types of testicular macrophage - important cells of the immune system - have been described in great detail by French researchers at the Centre d'Immunologie de Marseille-Luminy. (zmescience.com)
  • For example, macrophages present in the brain are termed microglia and in the liver sinusoids they are called Kupffer cells. (zmescience.com)
  • In the so-called interstitial space, where we can find the testosterone-producing Leydig cells, one type of interstitial macrophages is produced. (zmescience.com)
  • Macrophages are versatile cells that can be polarized by the tissue environment to fulfill required needs. (haematologica.org)
  • 6 In addition, these cells are present during early wound healing and proinflammatory macrophages are characterized by a pronounced ability to degrade tissue. (haematologica.org)
  • Macrophages secrete substances that attract other white blood cells to the site of the infection. (msdmanuals.com)
  • Taspine did not have an effect on specific assays for macrophage chemotaxis, neutrophil activation, fibroblast proliferation, or matrix assembly. (yourdictionary.com)
  • It is well-established that macrophages play an important role in many disease states but it has proven difficult to selectively target and alter macrophages that play a key role in disease progression," said Hagop Youssoufian, M.Sc. (businesswire.com)
  • Abnormal mononuclear macrophage infiltration may be involved in the origin and progression of CSD. (bmj.com)
  • Macrophages are known to be important in human muscle repair after acute damage. (the-scientist.com)
  • Start with ready-to-use, ethically sourced, primary human macrophages. (stemcell.com)
  • In human male testicles, scientists have identified two types of macrophages, each originating in one of the two compartments of the testis. (zmescience.com)
  • In contrast to monocytes, human macrophages did not show increased tissue factor expression upon stimulation with lipopolysaccharide and interferon-γ. (haematologica.org)
  • In summary, we will show that neuroimmunologists can learn many lessons from the well-developed field of macrophage phagocytosis biology. (frontiersin.org)
  • Vascular endothelial growth factor (VEGF), a key component in the process of angiogenesis, is known to be produced by tumour-associated macrophages 17 , 18 , which have been traditionally reported as M2 macrophages. (ersjournals.com)
  • All macrophages express common markers such as CD11b (Mac1 or CR3), CD68, and CD115 (M-CSF receptor). (hindawi.com)
  • The expression of CD68 for macrophages on different materials at 96 h. (figshare.com)
  • Despite extensive research, our understanding of the precise role of different subsets of macrophages in ischemia/reperfusion injury remains incomplete. (elifesciences.org)
  • However, how LRRK2 is activated in different macrophage subsets after membrane damage is unknown. (biorxiv.org)
  • These results suggest that LRRK2-regulated pathways of endolysosomal membrane damage and repair differ between macrophage subsets. (biorxiv.org)