A classification of lymphocytes based on structurally or functionally different populations of cells.
A classification of T-lymphocytes, especially into helper/inducer, suppressor/effector, and cytotoxic subsets, based on structurally or functionally different populations of cells.
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.
Ratio of T-LYMPHOCYTES that express the CD4 ANTIGEN to those that express the CD8 ANTIGEN. This value is commonly assessed in the diagnosis and staging of diseases affecting the IMMUNE SYSTEM including HIV INFECTIONS.
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.
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.
The number of LYMPHOCYTES per unit volume of BLOOD.
The number of WHITE BLOOD CELLS per unit volume in venous BLOOD. A differential leukocyte count measures the relative numbers of the different types of white cells.
A critical subpopulation of T-lymphocytes involved in the induction of most immunological functions. The HIV virus has selective tropism for the T4 cell which expresses the CD4 phenotypic marker, a receptor for HIV. In fact, the key element in the profound immunosuppression seen in HIV infection is the depletion of this subset of T-lymphocytes.
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.
Complex of at least five membrane-bound polypeptides in mature T-lymphocytes that are non-covalently associated with one another and with the T-cell receptor (RECEPTORS, ANTIGEN, T-CELL). The CD3 complex includes the gamma, delta, epsilon, zeta, and eta chains (subunits). When antigen binds to the T-cell receptor, the CD3 complex transduces the activating signals to the cytoplasm of the T-cell. The CD3 gamma and delta chains (subunits) are separate from and not related to the gamma/delta chains of the T-cell receptor (RECEPTORS, ANTIGEN, T-CELL, GAMMA-DELTA).
A critical subpopulation of regulatory T-lymphocytes involved in MHC Class I-restricted interactions. They include both cytotoxic T-lymphocytes (T-LYMPHOCYTES, CYTOTOXIC) and CD8+ suppressor T-lymphocytes.
Subpopulation of CD4+ lymphocytes that cooperate with other lymphocytes (either T or B) to initiate a variety of immune functions. For example, helper-inducer T-cells cooperate with B-cells to produce antibodies to thymus-dependent antigens and with other subpopulations of T-cells to initiate a variety of cell-mediated immune functions.
Process of classifying cells of the immune system based on structural and functional differences. The process is commonly used to analyze and sort T-lymphocytes into subsets based on CD antigens by the technique of flow cytometry.
Differentiation antigens found on thymocytes and on cytotoxic and suppressor T-lymphocytes. CD8 antigens are members of the immunoglobulin supergene family and are associative recognition elements in MHC (Major Histocompatibility Complex) Class I-restricted interactions.
CD4-positive T cells that inhibit immunopathology or autoimmune disease in vivo. They inhibit the immune response by influencing the activity of other cell types. Regulatory T-cells include naturally occurring CD4+CD25+ cells, IL-10 secreting Tr1 cells, and Th3 cells.
Antibodies produced by a single clone of cells.
High-molecular weight glycoproteins uniquely expressed on the surface of LEUKOCYTES and their hemopoietic progenitors. They contain a cytoplasmic protein tyrosine phosphatase activity which plays a role in intracellular signaling from the CELL SURFACE RECEPTORS. The CD45 antigens occur as multiple isoforms that result from alternative mRNA splicing and differential usage of three exons.
Bone marrow-derived lymphocytes that possess cytotoxic properties, classically directed against transformed and virus-infected cells. Unlike T CELLS; and B CELLS; NK CELLS are not antigen specific. The cytotoxicity of natural killer cells is determined by the collective signaling of an array of inhibitory and stimulatory CELL SURFACE RECEPTORS. A subset of T-LYMPHOCYTES referred to as NATURAL KILLER T CELLS shares some of the properties of this cell type.
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.
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.
An encapsulated lymphatic organ through which venous blood filters.
A subclass of HLA-D antigens that consist of alpha and beta chains. The inheritance of HLA-DR antigens differs from that of the HLA-DQ ANTIGENS and HLA-DP ANTIGENS.
T-cell receptors composed of CD3-associated gamma and delta polypeptide chains and expressed primarily in CD4-/CD8- T-cells. The receptors appear to be preferentially located in epithelial sites and probably play a role in the recognition of bacterial antigens. The T-cell receptor gamma/delta chains are separate and not related to the gamma and delta chains which are subunits of CD3 (see ANTIGENS, CD3).
Lymphoid cells concerned with humoral immunity. They are short-lived cells resembling bursa-derived lymphocytes of birds in their production of immunoglobulin upon appropriate stimulation.
Immunized T-lymphocytes which can directly destroy appropriate target cells. These cytotoxic lymphocytes may be generated in vitro in mixed lymphocyte cultures (MLC), in vivo during a graft-versus-host (GVH) reaction, or after immunization with an allograft, tumor cell or virally transformed or chemically modified target cell. The lytic phenomenon is sometimes referred to as cell-mediated lympholysis (CML). These CD8-positive cells are distinct from NATURAL KILLER CELLS and NATURAL KILLER T-CELLS. There are two effector phenotypes: TC1 and TC2.
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.
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.
A classification of B-lymphocytes based on structurally or functionally different populations of cells.
A soluble substance elaborated by antigen- or mitogen-stimulated T-LYMPHOCYTES which induces DNA synthesis in naive lymphocytes.
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.
Antigens expressed on the cell membrane of T-lymphocytes during differentiation, activation, and normal and neoplastic transformation. Their phenotypic characterization is important in differential diagnosis and studies of thymic ontogeny and T-cell function.
Mucoproteins isolated from the kidney bean (Phaseolus vulgaris); some of them are mitogenic to lymphocytes, others agglutinate all or certain types of erythrocytes or lymphocytes. They are used mainly in the study of immune mechanisms and in cell culture.
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.
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.
They are oval or bean shaped bodies (1 - 30 mm in diameter) located along the lymphatic system.
55-kDa antigens found on HELPER-INDUCER T-LYMPHOCYTES and on a variety of other immune cell types. CD4 antigens are members of the immunoglobulin supergene family and are implicated as associative recognition elements in MAJOR HISTOCOMPATIBILITY COMPLEX class II-restricted immune responses. On T-lymphocytes they define the helper/inducer subset. CD4 antigens also serve as INTERLEUKIN-15 receptors and bind to the HIV receptors, binding directly to the HIV ENVELOPE PROTEIN GP120.
Lymphocytes that show specificity for autologous tumor cells. Ex vivo isolation and culturing of TIL with interleukin-2, followed by reinfusion into the patient, is one form of adoptive immunotherapy of cancer.
Antigens on surfaces of cells, including infectious or foreign cells or viruses. They are usually protein-containing groups on cell membranes or walls and may be isolated.
A single, unpaired primary lymphoid organ situated in the MEDIASTINUM, extending superiorly into the neck to the lower edge of the THYROID GLAND and inferiorly to the fourth costal cartilage. It is necessary for normal development of immunologic function early in life. By puberty, it begins to involute and much of the tissue is replaced by fat.
A MANNOSE/GLUCOSE binding lectin isolated from the jack bean (Canavalia ensiformis). It is a potent mitogen used to stimulate cell proliferation in lymphocytes, primarily T-lymphocyte, cultures.
Measure of histocompatibility at the HL-A locus. Peripheral blood lymphocytes from two individuals are mixed together in tissue culture for several days. Lymphocytes from incompatible individuals will stimulate each other to proliferate significantly (measured by tritiated thymidine uptake) whereas those from compatible individuals will not. In the one-way MLC test, the lymphocytes from one of the individuals are inactivated (usually by treatment with MITOMYCIN or radiation) thereby allowing only the untreated remaining population of cells to proliferate in response to foreign histocompatibility antigens.
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.
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.
Molecules on the surface of T-lymphocytes that recognize and combine with antigens. The receptors are non-covalently associated with a complex of several polypeptides collectively called CD3 antigens (ANTIGENS, CD3). Recognition of foreign antigen and the major histocompatibility complex is accomplished by a single heterodimeric antigen-receptor structure, composed of either alpha-beta (RECEPTORS, ANTIGEN, T-CELL, ALPHA-BETA) or gamma-delta (RECEPTORS, ANTIGEN, T-CELL, GAMMA-DELTA) chains.
The in vitro formation of clusters consisting of a cell (usually a lymphocyte) surrounded by antigenic cells or antigen-bearing particles (usually erythrocytes, which may or may not be coated with antibody or antibody and complement). The rosette-forming cell may be an antibody-forming cell, a memory cell, a T-cell, a cell bearing surface cytophilic antibodies, or a monocyte possessing Fc receptors. Rosette formation can be used to identify specific populations of these cells.
Reduction in the number of lymphocytes.
Substances that stimulate mitosis and lymphocyte transformation. They include not only substances associated with LECTINS, but also substances from streptococci (associated with streptolysin S) and from strains of alpha-toxin-producing staphylococci. (Stedman, 25th ed)
A group of genetically identical cells all descended from a single common ancestral cell by mitosis in eukaryotes or by binary fission in prokaryotes. Clone cells also include populations of recombinant DNA molecules all carrying the same inserted sequence. (From King & Stansfield, Dictionary of Genetics, 4th ed)
Immunosuppression by reduction of circulating lymphocytes or by T-cell depletion of bone marrow. The former may be accomplished in vivo by thoracic duct drainage or administration of antilymphocyte serum. The latter is performed ex vivo on bone marrow before its transplantation.
The altered state of immunologic responsiveness resulting from initial contact with antigen, which enables the individual to produce antibodies more rapidly and in greater quantity in response to secondary antigenic stimulus.
Specialized tissues that are components of the lymphatic system. They provide fixed locations within the body where a variety of LYMPHOCYTES can form, mature and multiply. The lymphoid tissues are connected by a network of LYMPHATIC VESSELS.
A class of lymphocytes characterized by the lack of surface markers specific for either T or B lymphocytes.
The number of CD4-POSITIVE T-LYMPHOCYTES per unit volume of BLOOD. Determination requires the use of a fluorescence-activated flow cytometer.
T-cell receptors composed of CD3-associated alpha and beta polypeptide chains and expressed primarily in CD4+ or CD8+ T-cells. Unlike immunoglobulins, the alpha-beta T-cell receptors recognize antigens only when presented in association with major histocompatibility (MHC) molecules.
Receptors present on activated T-LYMPHOCYTES and B-LYMPHOCYTES that are specific for INTERLEUKIN-2 and play an important role in LYMPHOCYTE ACTIVATION. They are heterotrimeric proteins consisting of the INTERLEUKIN-2 RECEPTOR ALPHA SUBUNIT, the INTERLEUKIN-2 RECEPTOR BETA SUBUNIT, and the INTERLEUKIN RECEPTOR COMMON GAMMA-CHAIN.
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.
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.
Established cell cultures that have the potential to propagate indefinitely.
Sites on an antigen that interact with specific antibodies.
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.
Multi-subunit proteins which function in IMMUNITY. They are produced by B LYMPHOCYTES from the IMMUNOGLOBULIN GENES. They are comprised of two heavy (IMMUNOGLOBULIN HEAVY CHAINS) and two light chains (IMMUNOGLOBULIN LIGHT CHAINS) with additional ancillary polypeptide chains depending on their isoforms. The variety of isoforms include monomeric or polymeric forms, and transmembrane forms (B-CELL ANTIGEN RECEPTORS) or secreted forms (ANTIBODIES). They are divided by the amino acid sequence of their heavy chains into five classes (IMMUNOGLOBULIN A; IMMUNOGLOBULIN D; IMMUNOGLOBULIN E; IMMUNOGLOBULIN G; IMMUNOGLOBULIN M) and various subclasses.
A group of antigens that includes both the major and minor histocompatibility antigens. The former are genetically determined by the major histocompatibility complex. They determine tissue type for transplantation and cause allograft rejections. The latter are systems of allelic alloantigens that can cause weak transplant rejection.
Differentiation antigens expressed on B-lymphocytes and B-cell precursors. They are involved in regulation of B-cell proliferation.
Oligosaccharide antigenic determinants found principally on NK cells and T-cells. Their role in the immune response is poorly understood.
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.
The outward appearance of the individual. It is the product of interactions between genes, and between the GENOTYPE and the environment.
The demonstration of the cytotoxic effect on a target cell of a lymphocyte, a mediator released by a sensitized lymphocyte, an antibody, or complement.
Glycoproteins found on the membrane or surface of cells.
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).
Elements of limited time intervals, contributing to particular results or situations.
A CELL LINE derived from human T-CELL LEUKEMIA and used to determine the mechanism of differential susceptibility to anti-cancer drugs and radiation.
The type species of LENTIVIRUS and the etiologic agent of AIDS. It is characterized by its cytopathic effect and affinity for the T4-lymphocyte.
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.
The transfer of lymphocytes from a donor to a recipient or reinfusion to the donor.
T-cell enhancement of the B-cell response to thymic-dependent antigens.
IMMUNOGLOBULINS on the surface of B-LYMPHOCYTES. Their MESSENGER RNA contains an EXON with a membrane spanning sequence, producing immunoglobulins in the form of type I transmembrane proteins as opposed to secreted immunoglobulins (ANTIBODIES) which do not contain the membrane spanning segment.
Proteins isolated from the roots of the pokeweed, Phytolacca americana, that agglutinate some erythrocytes, stimulate mitosis and antibody synthesis in lymphocytes, and induce activation of plasma cells.
The 140 kDa isoform of NCAM (neural cell adhesion molecule) containing a transmembrane domain and short cytoplasmic tail. It is expressed by all lymphocytes mediating non-MHC restricted cytotoxicity and is present on some neural tissues and tumors.
The major immunoglobulin isotype class in normal human serum. There are several isotype subclasses of IgG, for example, IgG1, IgG2A, and IgG2B.
Glycoprotein members of the immunoglobulin superfamily which participate in T-cell adhesion and activation. They are expressed on most peripheral T-lymphocytes, natural killer cells, and thymocytes, and function as co-receptors or accessory molecules in the T-cell receptor complex.
The specific failure of a normally responsive individual to make an immune response to a known antigen. It results from previous contact with the antigen by an immunologically immature individual (fetus or neonate) or by an adult exposed to extreme high-dose or low-dose antigen, or by exposure to radiation, antimetabolites, antilymphocytic serum, etc.
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.
Costimulatory T-LYMPHOCYTE receptors that have specificity for CD80 ANTIGEN and CD86 ANTIGEN. Activation of this receptor results in increased T-cell proliferation, cytokine production and promotion of T-cell survival.
The fission of a CELL. It includes CYTOKINESIS, when the CYTOPLASM of a cell is divided, and CELL NUCLEUS DIVISION.
Progressive restriction of the developmental potential and increasing specialization of function that leads to the formation of specialized cells, tissues, and organs.
White blood cells. These include granular leukocytes (BASOPHILS; EOSINOPHILS; and NEUTROPHILS) as well as non-granular leukocytes (LYMPHOCYTES and MONOCYTES).
Includes the spectrum of human immunodeficiency virus infections that range from asymptomatic seropositivity, thru AIDS-related complex (ARC), to acquired immunodeficiency syndrome (AIDS).
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.
A class of immunoglobulin bearing mu chains (IMMUNOGLOBULIN MU-CHAINS). IgM can fix COMPLEMENT. The name comes from its high molecular weight and originally being called a macroglobulin.
A round-to-oval mass of lymphoid tissue embedded in the lateral wall of the PHARYNX. There is one on each side of the oropharynx in the fauces between the anterior and posterior pillars of the SOFT PALATE.
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.)
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.
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 production of ANTIBODIES by proliferating and differentiated B-LYMPHOCYTES under stimulation by ANTIGENS.
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.
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 movement of cells from one location to another. Distinguish from CYTOKINESIS which is the process of dividing the CYTOPLASM of a cell.
An integrin heterodimer widely expressed on cells of hematopoietic origin. CD11A ANTIGEN comprises the alpha chain and the CD18 antigen (ANTIGENS, CD18) the beta chain. Lymphocyte function-associated antigen-1 is a major receptor of T-CELLS; B-CELLS; and GRANULOCYTES. It mediates the leukocyte adhesion reactions underlying cytolytic conjugate formation, helper T-cell interactions, and antibody-dependent killing by NATURAL KILLER CELLS and granulocytes. Intracellular adhesion molecule-1 has been defined as a ligand for lymphocyte function-associated antigen-1.
Cell adhesion molecule and CD antigen that serves as a homing receptor for lymphocytes to lymph node high endothelial venules.
Cell surface glycoproteins on lymphocytes and other leukocytes that mediate adhesion to specialized blood vessels called high endothelial venules. Several different classes of lymphocyte homing receptors have been identified, and they appear to target different surface molecules (addressins) on high endothelial venules in different tissues. The adhesion plays a crucial role in the trafficking of lymphocytes.
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.
Laboratory mice that have been produced from a genetically manipulated EGG or EMBRYO, MAMMALIAN.
Syndromes in which there is a deficiency or defect in the mechanisms of immunity, either cellular or humoral.
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.
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.
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.
Antigenic determinants recognized and bound by the T-cell receptor. Epitopes recognized by the T-cell receptor are often located in the inner, unexposed side of the antigen, and become accessible to the T-cell receptors after proteolytic processing of the antigen.
The number of CELLS of a specific kind, usually measured per unit volume or area of sample.
The range or frequency distribution of a measurement in a population (of organisms, organs or things) that has not been selected for the presence of disease or abnormality.
Acquired defect of cellular immunity that occurs in cats infected with feline immunodeficiency virus (FIV) and in some cats infected with feline leukemia virus (FeLV).
Immunologic techniques based on the use of: (1) enzyme-antibody conjugates; (2) enzyme-antigen conjugates; (3) antienzyme antibody followed by its homologous enzyme; or (4) enzyme-antienzyme complexes. These are used histologically for visualizing or labeling tissue specimens.
'Rats, Inbred Lew' is a strain of laboratory rat that is widely used in biomedical research, known for its consistent genetic background and susceptibility to certain diseases, which makes it an ideal model for studying the genetic basis of complex traits and disease processes.
Proteins, glycoprotein, or lipoprotein moieties on surfaces of tumor cells that are usually identified by monoclonal antibodies. Many of these are of either embryonic or viral origin.
A specific HLA-A surface antigen subtype. Members of this subtype contain alpha chains that are encoded by the HLA-A*02 allele family.
Excess of normal lymphocytes in the blood or in any effusion.
The ability of lymphoid cells to mount a humoral or cellular immune response when challenged by antigen.
Glycoproteins expressed on all mature T-cells, thymocytes, and a subset of mature B-cells. Antibodies specific for CD5 can enhance T-cell receptor-mediated T-cell activation. The B-cell-specific molecule CD72 is a natural ligand for CD5. (From Abbas et al., Cellular and Molecular Immunology, 2d ed, p156)
Substances that are recognized by the immune system and induce an immune reaction.
The interstitial fluid that is in the LYMPHATIC SYSTEM.
Disorders that are characterized by the production of antibodies that react with host tissues or immune effector cells that are autoreactive to endogenous peptides.
The cells found in the body fluid circulating throughout the CARDIOVASCULAR SYSTEM.
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.
Form of passive immunization where previously sensitized immunologic agents (cells or serum) are transferred to non-immune recipients. When transfer of cells is used as a therapy for the treatment of neoplasms, it is called adoptive immunotherapy (IMMUNOTHERAPY, ADOPTIVE).
Subset of helper-inducer T-lymphocytes which synthesize and secrete interleukin-2, gamma-interferon, and interleukin-12. Due to their ability to kill antigen-presenting cells and their lymphokine-mediated effector activity, Th1 cells are associated with vigorous delayed-type hypersensitivity reactions.
Cell surface glycoproteins that bind to chemokines and thus mediate the migration of pro-inflammatory molecules. The receptors are members of the seven-transmembrane G protein-coupled receptor family. Like the CHEMOKINES themselves, the receptors can be divided into at least three structural branches: CR, CCR, and CXCR, according to variations in a shared cysteine motif.
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.
The process by which antigen is presented to lymphocytes in a form they can recognize. This is performed by antigen presenting cells (APCs). Some antigens require processing before they can be recognized. Antigen processing consists of ingestion and partial digestion of the antigen by the APC, followed by presentation of fragments on the cell surface. (From Rosen et al., Dictionary of Immunology, 1989)
An increased reactivity to specific antigens mediated not by antibodies but by cells.
A calcium-dependent pore-forming protein synthesized in cytolytic LYMPHOCYTES and sequestered in secretory granules. Upon immunological reaction between a cytolytic lymphocyte and a target cell, perforin is released at the plasma membrane and polymerizes into transmembrane tubules (forming pores) which lead to death of a target cell.
A worm-like blind tube extension from the CECUM.
Manipulation of the host's immune system in treatment of disease. It includes both active and passive immunization as well as immunosuppressive therapy to prevent graft rejection.
Transplantation between individuals of the same species. Usually refers to genetically disparate individuals in contradistinction to isogeneic transplantation for genetically identical individuals.
Serum containing GAMMA-GLOBULINS which are antibodies for lymphocyte ANTIGENS. It is used both as a test for HISTOCOMPATIBILITY and therapeutically in TRANSPLANTATION.
The major group of transplantation antigens in the mouse.
Membrane glycoproteins consisting of an alpha subunit and a BETA 2-MICROGLOBULIN beta subunit. In humans, highly polymorphic genes on CHROMOSOME 6 encode the alpha subunits of class I antigens and play an important role in determining the serological specificity of the surface antigen. Class I antigens are found on most nucleated cells and are generally detected by their reactivity with alloantisera. These antigens are recognized during GRAFT REJECTION and restrict cell-mediated lysis of virus-infected cells.
A heterogeneous group of immunocompetent cells that mediate the cellular immune response by processing and presenting antigens to the T-cells. Traditional antigen-presenting cells include MACROPHAGES; DENDRITIC CELLS; LANGERHANS CELLS; and B-LYMPHOCYTES. FOLLICULAR DENDRITIC CELLS are not traditional antigen-presenting cells, but because they hold antigen on their cell surface in the form of IMMUNE COMPLEXES for B-cell recognition they are considered so by some authors.
Adherence of cells to surfaces or to other cells.
Agents that suppress immune function by one of several mechanisms of action. Classical cytotoxic immunosuppressants act by inhibiting DNA synthesis. Others may act through activation of T-CELLS or by inhibiting the activation of HELPER CELLS. While immunosuppression has been brought about in the past primarily to prevent rejection of transplanted organs, new applications involving mediation of the effects of INTERLEUKINS and other CYTOKINES are emerging.
A group of closely related cyclic undecapeptides from the fungi Trichoderma polysporum and Cylindocarpon lucidum. They have some antineoplastic and antifungal action and significant immunosuppressive effects. Cyclosporins have been proposed as adjuvants in tissue and organ transplantation to suppress graft rejection.
Proteins prepared by recombinant DNA technology.
The sequence of PURINES and PYRIMIDINES in nucleic acids and polynucleotides. It is also called nucleotide sequence.
Proteins that share the common characteristic of binding to carbohydrates. Some ANTIBODIES and carbohydrate-metabolizing proteins (ENZYMES) also bind to carbohydrates, however they are not considered lectins. PLANT LECTINS are carbohydrate-binding proteins that have been primarily identified by their hemagglutinating activity (HEMAGGLUTININS). However, a variety of lectins occur in animal species where they serve diverse array of functions through specific carbohydrate recognition.
The body's defense mechanism against foreign organisms or substances and deviant native cells. It includes the humoral immune response and the cell-mediated response and consists of a complex of interrelated cellular, molecular, and genetic components.
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.
Surface ligands, usually glycoproteins, that mediate cell-to-cell adhesion. Their functions include the assembly and interconnection of various vertebrate systems, as well as maintenance of tissue integration, wound healing, morphogenic movements, cellular migrations, and metastasis.
All of the processes involved in increasing CELL NUMBER including CELL DIVISION.
Deliberate prevention or diminution of the host's immune response. It may be nonspecific as in the administration of immunosuppressive agents (drugs or radiation) or by lymphocyte depletion or may be specific as in desensitization or the simultaneous administration of antigen and immunosuppressive drugs.
Subset of helper-inducer T-lymphocytes which synthesize and secrete the interleukins IL-4, IL-5, IL-6, and IL-10. These cytokines influence B-cell development and antibody production as well as augmenting humoral responses.
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.
Lining of the INTESTINES, consisting of an inner EPITHELIUM, a middle LAMINA PROPRIA, and an outer MUSCULARIS MUCOSAE. In the SMALL INTESTINE, the mucosa is characterized by a series of folds and abundance of absorptive cells (ENTEROCYTES) with MICROVILLI.
An acquired defect of cellular immunity associated with infection by the human immunodeficiency virus (HIV), a CD4-positive T-lymphocyte count under 200 cells/microliter or less than 14% of total lymphocytes, and increased susceptibility to opportunistic infections and malignant neoplasms. Clinical manifestations also include emaciation (wasting) and dementia. These elements reflect criteria for AIDS as defined by the CDC in 1993.
Form of adoptive transfer where cells with antitumor activity are transferred to the tumor-bearing host in order to mediate tumor regression. The lymphoid cells commonly used are lymphokine-activated killer (LAK) cells and tumor-infiltrating lymphocytes (TIL). This is usually considered a form of passive immunotherapy. (From DeVita, et al., Cancer, 1993, pp.305-7, 314)
A method for the detection of very small quantities of antibody in which the antigen-antibody-complement complex adheres to indicator cells, usually primate erythrocytes or nonprimate blood platelets. The reaction is dependent on the number of bound C3 molecules on the C3b receptor sites of the indicator cell.
Proteins secreted from an organism which form membrane-spanning pores in target cells to destroy them. This is in contrast to PORINS and MEMBRANE TRANSPORT PROTEINS that function within the synthesizing organism and COMPLEMENT immune proteins. These pore forming cytotoxic proteins are a form of primitive cellular defense which are also found in human LYMPHOCYTES.
Histochemical localization of immunoreactive substances using labeled antibodies as reagents.
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.
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.
Either of the pair of organs occupying the cavity of the thorax that effect the aeration of the blood.
A family of serine endopeptidases found in the SECRETORY GRANULES of LEUKOCYTES such as CYTOTOXIC T-LYMPHOCYTES and NATURAL KILLER CELLS. When secreted into the intercellular space granzymes act to eliminate transformed and virus-infected host cells.
Disease having a short and relatively severe course.
A malignant neoplasm derived from cells that are capable of forming melanin, which may occur in the skin of any part of the body, in the eye, or, rarely, in the mucous membranes of the genitalia, anus, oral cavity, or other sites. It occurs mostly in adults and may originate de novo or from a pigmented nevus or malignant lentigo. Melanomas frequently metastasize widely, and the regional lymph nodes, liver, lungs, and brain are likely to be involved. The incidence of malignant skin melanomas is rising rapidly in all parts of the world. (Stedman, 25th ed; from Rook et al., Textbook of Dermatology, 4th ed, p2445)
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.
Cells grown in vitro from neoplastic tissue. If they can be established as a TUMOR CELL LINE, they can be propagated in cell culture indefinitely.
A member of the tumor necrosis factor receptor superfamily found on most T-LYMPHOCYTES. Activation of the receptor by CD70 ANTIGEN results in the increased proliferation of CD4-POSITIVE T-LYMPHOCYTES and CD8-POSITIVE T-LYMPHOCYTES. Signaling by the activated receptor occurs through its association with TNF RECEPTOR-ASSOCIATED FACTORS.
Leukemia associated with HYPERPLASIA of the lymphoid tissues and increased numbers of circulating malignant LYMPHOCYTES and lymphoblasts.
A tumor necrosis factor receptor subtype found in a variety of tissues and on activated LYMPHOCYTES. It has specificity for FAS LIGAND and plays a role in regulation of peripheral immune responses and APOPTOSIS. Multiple isoforms of the protein exist due to multiple ALTERNATIVE SPLICING. The activated receptor signals via a conserved death domain that associates with specific TNF RECEPTOR-ASSOCIATED FACTORS in the CYTOPLASM.
A species of LENTIVIRUS, subgenus feline lentiviruses (LENTIVIRUSES, FELINE) isolated from cats with a chronic wasting syndrome, presumed to be immune deficiency. There are 3 strains: Petaluma (FIP-P), Oma (FIP-O) and Puma lentivirus (PLV). There is no antigenic relationship between FIV and HIV, nor does FIV grow in human T-cells.
A transmembrane protein belonging to the tumor necrosis factor superfamily that was originally discovered on cells of the lymphoid-myeloid lineage, including activated T-LYMPHOCYTES and NATURAL KILLER CELLS. It plays an important role in immune homeostasis and cell-mediated toxicity by binding to the FAS RECEPTOR and triggering APOPTOSIS.
A low affinity interleukin-2 receptor subunit that combines with the INTERLEUKIN-2 RECEPTOR BETA SUBUNIT and the INTERLEUKIN RECEPTOR COMMON GAMMA-CHAIN to form a high affinity receptor for INTERLEUKIN-2.
Unglycosylated phosphoproteins expressed only on B-cells. They are regulators of transmembrane Ca2+ conductance and thought to play a role in B-cell activation and proliferation.
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.
Blood of the fetus. Exchange of nutrients and waste between the fetal and maternal blood occurs via the PLACENTA. The cord blood is blood contained in the umbilical vessels (UMBILICAL CORD) at the time of delivery.
The genetic region which contains the loci of genes which determine the structure of the serologically defined (SD) and lymphocyte-defined (LD) TRANSPLANTATION ANTIGENS, genes which control the structure of the IMMUNE RESPONSE-ASSOCIATED ANTIGENS, HUMAN; the IMMUNE RESPONSE GENES which control the ability of an animal to respond immunologically to antigenic stimuli, and genes which determine the structure and/or level of the first four components of complement.
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.
Substances elaborated by viruses that have antigenic activity.
Nonsusceptibility to the invasive or pathogenic effects of foreign microorganisms or to the toxic effect of antigenic substances.
The movement of leukocytes in response to a chemical concentration gradient or to products formed in an immunologic reaction.
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.
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.
Any of the ruminant mammals with curved horns in the genus Ovis, family Bovidae. They possess lachrymal grooves and interdigital glands, which are absent in GOATS.
The rate dynamics in chemical or physical systems.
Studies which start with the identification of persons with a disease of interest and a control (comparison, referent) group without the disease. The relationship of an attribute to the disease is examined by comparing diseased and non-diseased persons with regard to the frequency or levels of the attribute in each group.
The outer covering of the body that protects it from the environment. It is composed of the DERMIS and the EPIDERMIS.
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.
Antigens determined by leukocyte loci found on chromosome 6, the major histocompatibility loci in humans. They are polypeptides or glycoproteins found on most nucleated cells and platelets, determine tissue types for transplantation, and are associated with certain diseases.
A specific immune response elicited by a specific dose of an immunologically active substance or cell in an organism, tissue, or cell.
A species of the genus MACACA inhabiting India, China, and other parts of Asia. The species is used extensively in biomedical research and adapts very well to living with humans.
Polymorphic class I human histocompatibility (HLA) surface antigens present on almost all nucleated cells. At least 20 antigens have been identified which are encoded by the A locus of multiple alleles on chromosome 6. They serve as targets for T-cell cytolytic responses and are involved with acceptance or rejection of tissue/organ grafts.
Deliberate stimulation of the host's immune response. ACTIVE IMMUNIZATION involves administration of ANTIGENS or IMMUNOLOGIC ADJUVANTS. PASSIVE IMMUNIZATION involves administration of IMMUNE SERA or LYMPHOCYTES or their extracts (e.g., transfer factor, immune RNA) or transplantation of immunocompetent cell producing tissue (thymus or bone marrow).
An idiopathic systemic inflammatory granulomatous disorder comprised of epithelioid and multinucleated giant cells with little necrosis. It usually invades the lungs with fibrosis and may also involve lymph nodes, skin, liver, spleen, eyes, phalangeal bones, and parotid glands.
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.
The property of antibodies which enables them to react with some ANTIGENIC DETERMINANTS and not with others. Specificity is dependent on chemical composition, physical forces, and molecular structure at the binding site.
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.
Progenitor cells from which all blood cells derive.
The transference of BONE MARROW from one human or animal to another for a variety of purposes including HEMATOPOIETIC STEM CELL TRANSPLANTATION or MESENCHYMAL STEM CELL TRANSPLANTATION.
A heterogeneous group of infections produced by coxsackieviruses, including HERPANGINA, aseptic meningitis (MENINGITIS, ASEPTIC), a common-cold-like syndrome, a non-paralytic poliomyelitis-like syndrome, epidemic pleurodynia (PLEURODYNIA, EPIDEMIC) and a serious MYOCARDITIS.
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).
The gradual irreversible changes in structure and function of an organism that occur as a result of the passage of time.
Represents 15-20% of the human serum immunoglobulins, mostly as the 4-chain polymer in humans or dimer in other mammals. Secretory IgA (IMMUNOGLOBULIN A, SECRETORY) is the main immunoglobulin in secretions.
A membrane-bound or cytosolic enzyme that catalyzes the synthesis of CYCLIC ADP-RIBOSE (cADPR) from nicotinamide adenine dinucleotide (NAD). This enzyme generally catalyzes the hydrolysis of cADPR to ADP-RIBOSE, as well, and sometimes the synthesis of cyclic ADP-ribose 2' phosphate (2'-P-cADPR) from NADP.
Antibodies that react with self-antigens (AUTOANTIGENS) of the organism that produced them.

Crystal structure of an MHC class I presented glycopeptide that generates carbohydrate-specific CTL. (1/8823)

T cell receptor (TCR) recognition of nonpeptidic and modified peptide antigens has been recently uncovered but is still poorly understood. Immunization with an H-2Kb-restricted glycopeptide RGY8-6H-Gal2 generates a population of cytotoxic T cells that express both alpha/beta TCR, specific for glycopeptide, and gamma/delta TCR, specific for the disaccharide, even on glycolipids. The crystal structure of Kb/RGY8-6H-Gal2 now demonstrates that the peptide and H-2Kb structures are unaffected by the peptide glycosylation, but the central region of the putative TCR binding site is dominated by the extensive exposure of the tethered carbohydrate. These features of the Kb/RGY8-6H-Gal2 structure are consistent with the individual ligand binding preferences identified for the alpha/beta and gamma/delta TCRs and thus explain the generation of a carbohydrate-specific T cell response.  (+info)

T cell subsets in experimental lupus nephritis: modulation by bacterial superantigen. (2/8823)

Chronic graft-vs-host disease (GvH), induced by injection of DBA/2 lymphocytes into (C57BL/6 x DBA/2)F1 hybrids, is a murine model for lupus nephritis, associated with a Th2-dependent polyclonal B cell activation. The development of glomerulosclerosis in this model is preceded by a glomerular influx of LFA-1+ T cells. We investigated whether exposure to bacterial superantigen would modulate the course of this autoimmune syndrome. Injection of the bacterial superantigen staphylococcal enterotoxin B (SEB) in mice has been shown to induce the activation of TcRVbeta8+ T cells. Within 2 weeks after GvH induction, mice were injected twice with 20 microg of SEB and the following parameters were examined: cytokine and Ig profile, proteinuria and renal pathology. The second SEB injection induced in GvH mice an increased release of both interferon-gamma (IFN-gamma) and interleukin-10 (IL-10) as compared with control F1 mice. No differences were observed in IL-2 production. SEB-treated GvH mice demonstrated a delayed onset of proteinuria. Histological analysis of the kidney showed that SEB-challenged GvH mice displayed significantly more interstitial inflammation and mesangial proliferation together with more IgG2a deposits in glomeruli than non-injected GvH mice. From these results, we conclude that GvH mice are more responsive to SEB in terms of cytokine production and that bacterial infection can modulate the course of this renal disease from a membranous to a more proliferative type of nephropathy.  (+info)

Melanoma cells present a MAGE-3 epitope to CD4(+) cytotoxic T cells in association with histocompatibility leukocyte antigen DR11. (3/8823)

In this study we used TEPITOPE, a new epitope prediction software, to identify sequence segments on the MAGE-3 protein with promiscuous binding to histocompatibility leukocyte antigen (HLA)-DR molecules. Synthetic peptides corresponding to the identified sequences were synthesized and used to propagate CD4(+) T cells from the blood of a healthy donor. CD4(+) T cells strongly recognized MAGE-3281-295 and, to a lesser extent, MAGE-3141-155 and MAGE-3146-160. Moreover, CD4(+) T cells proliferated in the presence of recombinant MAGE-3 after processing and presentation by autologous antigen presenting cells, demonstrating that the MAGE-3 epitopes recognized are naturally processed. CD4(+) T cells, mostly of the T helper 1 type, showed specific lytic activity against HLA-DR11/MAGE-3-positive melanoma cells. Cold target inhibition experiments demonstrated indeed that the CD4(+) T cells recognized MAGE-3281-295 in association with HLA-DR11 on melanoma cells. This is the first evidence that a tumor-specific shared antigen forms CD4(+) T cell epitopes. Furthermore, we validated the use of algorithms for the prediction of promiscuous CD4(+) T cell epitopes, thus opening the possibility of wide application to other tumor-associated antigens. These results have direct implications for cancer immunotherapy in the design of peptide-based vaccines with tumor-specific CD4(+) T cell epitopes.  (+info)

Peripheral autoantigen induces regulatory T cells that prevent autoimmunity. (4/8823)

Previous studies have shown that autoimmune thyroiditis can be induced in normal laboratory rats after thymectomy and split dose gamma-irradiation. Development of disease can be prevented by reconstitution of PVG rats shortly after their final irradiation with either peripheral CD4(+)CD45RC- T cells or CD4(+)CD8(-) thymocytes from syngeneic donors. Although the activity of both populations is known to depend on the activities of endogenously produced interleukin 4 and transforming growth factor beta, implying a common mechanism, the issue of antigen specificity of the cells involved has not yet been addressed. In this study, we show that the regulatory T cells that prevent autoimmune thyroiditis are generated in vivo only when the relevant autoantigen is also present. Peripheral CD4(+) T cells, from rats whose thyroids were ablated in utero by treatment with 131I, were unable to prevent disease development upon adoptive transfer into thymectomized and irradiated recipients. This regulatory deficit is specific for thyroid autoimmunity, since CD4(+) T cells from 131I-treated PVG.RT1(u) rats were as effective as those from normal donors at preventing diabetes in thymectomized and irradiated PVG.RT1(u) rats. Significantly, in contrast to the peripheral CD4(+) T cells, CD4(+)CD8(-) thymocytes from 131I-treated PVG donors were still able to prevent thyroiditis upon adoptive transfer. Taken together, these data indicate that it is the peripheral autoantigen itself that stimulates the generation of the appropriate regulatory cells from thymic emigrant precursors.  (+info)

Selective recruitment of CCR4-bearing Th2 cells toward antigen-presenting cells by the CC chemokines thymus and activation-regulated chemokine and macrophage-derived chemokine. (5/8823)

Helper T cells are classified into Th1 and Th2 subsets based on their profiles of cytokine production. Th1 cells are involved in cell-mediated immunity, whereas Th2 cells induce humoral responses. Selective recruitment of these two subsets depends on specific adhesion molecules and specific chemoattractants. Here, we demonstrate that the T cell-directed CC chemokine thymus and activation-regulated chemokine (TARC) was abundantly produced by monocytes treated with granulocyte macrophage colony stimulating factor (GM-CSF) or IL-3, especially in the presence of IL-4 and by dendritic cells derived from monocytes cultured with GM-CSF + IL-4. The receptor for TARC and another macrophage/dendritic cell-derived CC chemokine macrophage-derived chemokine (MDC) is CCR4, a G protein-coupled receptor. CCR4 was found to be expressed on approximately 20% of adult peripheral blood effector/memory CD4+ T cells. T cells attracted by TARC and MDC generated cell lines predominantly producing Th2-type cytokines, IL-4 and IL-5. Fractionated CCR4+ cells but not CCR4- cells also selectively gave rise to Th2-type cell lines. When naive CD4+ T cells from adult peripheral blood were polarized in vitro, Th2-type cells selectively expressed CCR4 and vigorously migrated toward TARC and MDC. Taken together, CCR4 is selectively expressed on Th2-type T cells and antigen-presenting cells may recruit Th2 cells expressing CCR4 by producing TARC and MDC in Th2-dominant conditions.  (+info)

Development and function of autospecific dual TCR+ T lymphocytes. (6/8823)

Recent studies have challenged the long held concept that each T lymphocyte expresses on its surface only a single, unique alphabetaTCR. Dual TCR+ T cells have been recognized, however, their origin and potential to escape screening for self-reactivity remain obscure. We now report the thymic generation of dual alphabetaTCR+ T cells in the H-2Db/H-Y-specific TCR transgenic (Tg) mouse. Dual TCR+ thymocytes were positively selected less efficiently than single TCR+ thymocytes, although a subset attained maturity. Importantly, when TCR Tg mice were bred onto a negatively selecting background, auto-specific cells survived central deletion and matured as CD4+ dual TCR+ cells. These cells were autoreactive when CD8 expression was restored. The existence of autospecific, dual TCR+ T cells may have implications for the maintenance of self tolerance.  (+info)

Patterns of A2A extracellular adenosine receptor expression in different functional subsets of human peripheral T cells. Flow cytometry studies with anti-A2A receptor monoclonal antibodies. (7/8823)

Signaling through A2A adenosine receptors (A2AR) regulates T lymphocyte expansion and modulates T cell receptor (TCR)-mediated effector functions in vitro. To understand the role of A2ARs in the regulation of immune response, we investigated the expression levels of this receptor in different functional lymphocyte subsets. Monoclonal anti-A2AR antibody was used to develop a flow cytometric assay to quantify the expression A2ARs on lymphocytes. We report that detectable levels of expression of A2ARs are much higher among T cells than B cells. More CD4(+) than CD8(+) T cells express A2ARs, but activation of T cells increases A2AR expression, predominantly in CD8(+) T cells. No significant differences were found in the proportion of A2AR+ cells between CD8(low) and CD8(high) T cells or between TCR/CD3(low) and TCR/CD3(high) T cells. Studies of T helper cell subsets (TH1 and TH2) reveal that lymphokine-producing cells are much more likely to express A2ARs than are cells that do not produce lymphokines. These results suggest that A2ARs are variably expressed on T cell subsets and may regulate cytokine production in activated T lymphocytes.  (+info)

IgA production in MHC class II-deficient mice is primarily a function of B-1a cells. (8/8823)

Mice deficient in MHC class II expression (C2d mice) do not make antibody to protein antigens administered systemically, but their ability to produce IgA antibody to antigen administered at mucosal sites has not been described. We investigated IgA production by C2d mice and their IgA antibody response to antigen given orally. Young C2d mice had normal amounts of serum IgA, intestinal-secreted IgA and normal numbers of intestinal IgA plasma cells, compared to control C57BL/6 mice. IgA production by C2d mice increased with age. Following oral immunization with cholera toxin, C57BL/6 mice responded with IgA and IgG antibody, and had increased numbers of IgA plasma cells, but C2d mice gave no response. The Peyer's patch and mesenteric lymph node tissues of C2d mice contained very few CD4-expressing T cells. Thus, C2d mice have no typical mucosal CD4 Th cells and cannot respond to a strong oral immunogen, yet they still produced and secreted IgA. We hypothesized that B-1 lymphocytes could provide a source of IgA independent of antigen-specific T cell help. Young C2d mice had normal numbers of peritoneal B-1a cells and their frequency increased with age. To test the role of these B-1a cells, we bred C2d mice to obtain mice that had no MHC class II expression and expressed the Xid gene that confers deficiency in B-1a cells. These double-deficient mice had 10-fold less serum and secreted IgA than all other F2 littermates. We conclude that B-1a cells are essential for the majority of IgA production in C2d mice. Thus, the C2d mouse may provide a useful tool for analysis of the role of intestinal IgA provided by B-1a cells.  (+info)

Lymphocyte subsets refer to distinct populations of white blood cells called lymphocytes, which are crucial components of the adaptive immune system. There are two main types of lymphocytes: T cells and B cells, and each type has several subsets based on their surface receptors, functions, and activation status.

1. T cell subsets: These include CD4+ T helper cells (Th cells), CD8+ cytotoxic T cells (Tc cells), regulatory T cells (Tregs), and memory T cells. Th cells are further divided into Th1, Th2, Th17, and Tfh cells based on their cytokine production profiles and functions.
* CD4+ T helper cells (Th cells) play a central role in orchestrating the immune response by producing various cytokines that activate other immune cells.
* CD8+ cytotoxic T cells (Tc cells) directly kill virus-infected or malignant cells upon recognition of specific antigens presented on their surface.
* Regulatory T cells (Tregs) suppress the activation and proliferation of other immune cells to maintain self-tolerance and prevent autoimmunity.
* Memory T cells are long-lived cells that remain in the body after an initial infection or immunization, providing rapid protection upon subsequent encounters with the same pathogen.
2. B cell subsets: These include naïve B cells, memory B cells, and plasma cells. Upon activation by antigens, B cells differentiate into antibody-secreting plasma cells that produce specific antibodies to neutralize or eliminate pathogens.
* Naïve B cells are resting cells that have not yet encountered their specific antigen.
* Memory B cells are long-lived cells generated after initial antigen exposure, which can quickly differentiate into antibody-secreting plasma cells upon re-exposure to the same antigen.
* Plasma cells are terminally differentiated B cells that secrete large amounts of specific antibodies.

Analyzing lymphocyte subsets is essential for understanding immune system function and dysfunction, as well as monitoring the effectiveness of immunotherapies and vaccinations.

T-lymphocyte subsets refer to distinct populations of T-cells, which are a type of white blood cell that plays a central role in cell-mediated immunity. The two main types of T-lymphocytes are CD4+ and CD8+ cells, which are defined by the presence or absence of specific proteins called cluster differentiation (CD) molecules on their surface.

CD4+ T-cells, also known as helper T-cells, play a crucial role in activating other immune cells, such as B-lymphocytes and macrophages, to mount an immune response against pathogens. They also produce cytokines that help regulate the immune response.

CD8+ T-cells, also known as cytotoxic T-cells, directly kill infected cells or tumor cells by releasing toxic substances such as perforins and granzymes.

The balance between these two subsets of T-cells is critical for maintaining immune homeostasis and mounting effective immune responses against pathogens while avoiding excessive inflammation and autoimmunity. Therefore, the measurement of T-lymphocyte subsets is essential in diagnosing and monitoring various immunological disorders, including HIV infection, cancer, and autoimmune diseases.

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.

The CD4-CD8 ratio is a measurement of the relative numbers of two types of immune cells, CD4+ T cells (also known as helper T cells) and CD8+ T cells (also known as cytotoxic T cells), in the blood. The CD4-CD8 ratio is commonly used as a marker of immune function and health.

CD4+ T cells play an important role in the immune response by helping to coordinate the activity of other immune cells, producing chemical signals that activate them, and producing antibodies. CD8+ T cells are responsible for directly killing infected cells and tumor cells.

A normal CD4-CD8 ratio is typically between 1.0 and 3.0. A lower ratio may indicate an impaired immune system, such as in cases of HIV infection or other immunodeficiency disorders. A higher ratio may be seen in some viral infections, autoimmune diseases, or cancer. It's important to note that the CD4-CD8 ratio should be interpreted in conjunction with other laboratory and clinical findings for a more accurate assessment of immune function.

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.

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.

A lymphocyte count is a laboratory test that measures the number of white blood cells called lymphocytes in a sample of blood. Lymphocytes are a vital part of the immune system and help fight off infections and diseases. A normal lymphocyte count ranges from 1,000 to 4,800 cells per microliter (µL) of blood for adults.

An abnormal lymphocyte count can indicate an infection, immune disorder, or blood cancer. A low lymphocyte count is called lymphopenia, while a high lymphocyte count is called lymphocytosis. The cause of an abnormal lymphocyte count should be investigated through further testing and clinical evaluation.

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.

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.

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.

CD3 antigens are a group of proteins found on the surface of T-cells, which are a type of white blood cell that plays a central role in the immune response. The CD3 antigens are composed of several different subunits (ε, δ, γ, and α) that associate to form the CD3 complex, which is involved in T-cell activation and signal transduction.

The CD3 complex is associated with the T-cell receptor (TCR), which recognizes and binds to specific antigens presented by antigen-presenting cells. When the TCR binds to an antigen, it triggers a series of intracellular signaling events that lead to T-cell activation and the initiation of an immune response.

CD3 antigens are important targets for immunotherapy in some diseases, such as certain types of cancer. For example, monoclonal antibodies that target CD3 have been developed to activate T-cells and enhance their ability to recognize and destroy tumor cells. However, CD3-targeted therapies can also cause side effects, such as cytokine release syndrome, which can be serious or life-threatening in some cases.

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.

T-lymphocytes, also known as T-cells, are a type of white blood cell that plays a key role in the immune response. They help to protect the body from infection and disease by identifying and attacking foreign substances such as viruses and bacteria.

Helper-inducer T-lymphocytes, also known as CD4+ T-cells or Th0 cells, are a specific subset of T-lymphocytes that help to coordinate the immune response. They do this by activating other immune cells, such as B-lymphocytes (which produce antibodies) and cytotoxic T-lymphocytes (which directly attack infected cells). Helper-inducer T-lymphocytes also release cytokines, which are signaling molecules that help to regulate the immune response.

Helper-inducer T-lymphocytes can differentiate into different subsets of T-cells, depending on the type of cytokines they are exposed to. For example, they can differentiate into Th1 cells, which produce cytokines that help to activate cytotoxic T-lymphocytes and macrophages; or Th2 cells, which produce cytokines that help to activate B-lymphocytes and eosinophils.

It is important to note that helper-inducer T-lymphocytes play a crucial role in the immune response, and dysfunction of these cells can lead to immunodeficiency or autoimmune disorders.

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.

CD8 antigens are a type of protein found on the surface of certain immune cells called cytotoxic T lymphocytes or cytotoxic T cells. These cells play a critical role in the adaptive immune response, which is the specific and targeted response of the immune system to foreign substances (antigens) that invade the body.

CD8 antigens help cytotoxic T cells recognize and respond to infected or abnormal cells, such as those that have been infected by a virus or have become cancerous. When a cytotoxic T cell encounters a cell displaying a specific antigen bound to a CD8 molecule, it becomes activated and releases toxic substances that can kill the target cell.

CD8 antigens are also known as cluster of differentiation 8 antigens or CD8 receptors. They belong to a larger family of proteins called major histocompatibility complex class I (MHC class I) molecules, which present antigens to T cells and play a crucial role in the immune system's ability to distinguish between self and non-self.

Regulatory T-lymphocytes (Tregs), also known as suppressor T cells, are a subpopulation of T-cells that play a critical role in maintaining immune tolerance and preventing autoimmune diseases. They function to suppress the activation and proliferation of other immune cells, thereby regulating the immune response and preventing it from attacking the body's own tissues.

Tregs constitutively express the surface markers CD4 and CD25, as well as the transcription factor Foxp3, which is essential for their development and function. They can be further divided into subsets based on their expression of other markers, such as CD127 and CD45RA.

Tregs are critical for maintaining self-tolerance by suppressing the activation of self-reactive T cells that have escaped negative selection in the thymus. They also play a role in regulating immune responses to foreign antigens, such as those encountered during infection or cancer, and can contribute to the immunosuppressive microenvironment found in tumors.

Dysregulation of Tregs has been implicated in various autoimmune diseases, including type 1 diabetes, rheumatoid arthritis, and multiple sclerosis, as well as in cancer and infectious diseases. Therefore, understanding the mechanisms that regulate Treg function is an important area of research with potential therapeutic implications.

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.

CD45 is a protein that is found on the surface of many types of white blood cells, including T-cells, B-cells, and natural killer (NK) cells. It is also known as leukocyte common antigen because it is present on almost all leukocytes. CD45 is a tyrosine phosphatase that plays a role in regulating the activity of various proteins involved in cell signaling pathways.

As an antigen, CD45 is used as a marker to identify and distinguish different types of white blood cells. It has several isoforms that are generated by alternative splicing of its mRNA, resulting in different molecular weights. The size of the CD45 isoform can be used to distinguish between different subsets of T-cells and B-cells.

CD45 is an important molecule in the immune system, and abnormalities in its expression or function have been implicated in various diseases, including autoimmune disorders and cancer.

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.

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.

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.

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.

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.

1. Receptors: In the context of physiology and medicine, receptors are specialized proteins found on the surface of cells or inside cells that detect and respond to specific molecules, known as ligands. They play a crucial role in various biological processes, including signal transduction, cell communication, and regulation of physiological functions.
2. Antigen: An antigen is a foreign substance (usually a protein) that triggers an immune response when introduced into the body. Antigens can be derived from various sources, such as bacteria, viruses, fungi, or parasites. They are recognized by the immune system as non-self and stimulate the production of antibodies and activation of immune cells, like T-cells, to eliminate the threat.
3. T-Cell: T-cells, also known as T-lymphocytes, are a type of white blood cell that plays a central role in cell-mediated immunity. They are produced in the bone marrow and mature in the thymus gland. T-cells have receptors on their surface called T-cell receptors (TCRs) that enable them to recognize and respond to specific antigens presented by antigen-presenting cells (APCs). There are several types of T-cells, including CD4+ helper T-cells, CD8+ cytotoxic T-cells, and regulatory T-cells.
4. gamma-delta (γδ) T-Cell: Gamma-delta (γδ) T-cells are a subset of T-cells that possess a distinct T-cell receptor (TCR) composed of gamma and delta chains. Unlike conventional T-cells, which typically recognize peptide antigens presented by major histocompatibility complex (MHC) molecules, γδ T-cells can directly recognize various non-peptide antigens, such as lipids, glycolipids, and small metabolites. They are involved in the early stages of immune responses, tissue homeostasis, and cancer surveillance.

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.

Cytotoxic T-lymphocytes, also known as CD8+ T cells, are a type of white blood cell that plays a central role in the cell-mediated immune system. They are responsible for identifying and destroying virus-infected cells and cancer cells. When a cytotoxic T-lymphocyte recognizes a specific antigen presented on the surface of an infected or malignant cell, it becomes activated and releases toxic substances such as perforins and granzymes, which can create pores in the target cell's membrane and induce apoptosis (programmed cell death). This process helps to eliminate the infected or malignant cells and prevent the spread of infection or cancer.

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.

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.

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

B-lymphocyte subsets refer to distinct populations of B-cells that can be identified based on their surface receptors and functional characteristics. Some common B-lymphocyte subsets include:

1. Naive B-cells: These are mature B-cells that have not yet been exposed to an antigen. They express surface receptors called immunoglobulin M (IgM) and immunoglobulin D (IgD).
2. Memory B-cells: These are B-cells that have previously encountered an antigen and mounted an immune response. They express high levels of surface immunoglobulins and can quickly differentiate into antibody-secreting plasma cells upon re-exposure to the same antigen.
3. Plasma cells: These are fully differentiated B-cells that secrete large amounts of antibodies in response to an antigen. They lack surface immunoglobulins and do not undergo further division.
4. Regulatory B-cells: These are a subset of B-cells that modulate the immune response by producing anti-inflammatory cytokines and suppressing the activation of other immune cells.
5. B-1 cells: These are a population of B-cells that are primarily found in the peripheral blood and mucosal tissues. They produce natural antibodies that provide early protection against pathogens and help to maintain tissue homeostasis.

Understanding the different B-lymphocyte subsets and their functions is important for diagnosing and treating immune-related disorders, including autoimmune diseases, infections, and cancer.

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.

"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.

Antigens are substances (usually proteins) on the surface of cells, viruses, fungi, or bacteria that the immune system recognizes as foreign and mounts a response against.

Differentiation in the context of T-lymphocytes refers to the process by which immature T-cells mature and develop into different types of T-cells with specific functions, such as CD4+ helper T-cells or CD8+ cytotoxic T-cells.

T-lymphocytes, also known as T-cells, are a type of white blood cell that plays a central role in cell-mediated immunity. They are produced in the bone marrow and mature in the thymus gland. Once mature, they circulate throughout the body in search of foreign antigens to attack and destroy.

Therefore, 'Antigens, Differentiation, T-Lymphocyte' refers to the process by which T-lymphocytes mature and develop the ability to recognize and respond to specific foreign antigens.

Phytohemagglutinins (PHA) are a type of lectin, specifically a mitogen, found in certain plants such as red kidney beans, white kidney beans, and butter beans. They have the ability to agglutinate erythrocytes (red blood cells) and stimulate the proliferation of lymphocytes (a type of white blood cell). PHA is often used in medical research and diagnostics as a means to study immune system function, particularly the activation and proliferation of T-cells. It's also used in some immunological assays. However, it should be noted that ingesting large amounts of raw or undercooked beans containing high levels of PHA can cause adverse gastrointestinal symptoms due to their ability to interact with the cells lining the digestive tract.

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.

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.

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.

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.

Tumor-infiltrating lymphocytes (TILs) are a type of immune cell that have migrated from the bloodstream into a tumor. They are primarily composed of T cells, B cells, and natural killer (NK) cells. TILs can be found in various types of solid tumors, and their presence and composition have been shown to correlate with patient prognosis and response to certain therapies.

TILs play a crucial role in the immune response against cancer, as they are able to recognize and kill cancer cells. They can also release cytokines and chemokines that attract other immune cells to the tumor site, enhancing the anti-tumor immune response. However, tumors can develop mechanisms to evade or suppress the immune response, including the suppression of TILs.

TILs have emerged as a promising target for cancer immunotherapy, with adoptive cell transfer (ACT) being one of the most widely studied approaches. In ACT, TILs are isolated from a patient's tumor, expanded in the laboratory, and then reinfused back into the patient to enhance their anti-tumor immune response. This approach has shown promising results in clinical trials for several types of cancer, including melanoma and cervical cancer.

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.

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.

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.

A Lymphocyte Culture Test, Mixed (LCTM) is not a standardized medical test with a universally accepted definition. However, in some contexts, it may refer to a laboratory procedure where both T-lymphocytes and B-lymphocytes are cultured together from a sample of peripheral blood or other tissues. This test is sometimes used in research or specialized diagnostic settings to evaluate the immune function or to study the interactions between T-cells and B-cells in response to various stimuli, such as antigens or mitogens.

The test typically involves isolating lymphocytes from a sample, adding them to a culture medium along with appropriate stimulants, and then incubating the mixture for a period of time. The resulting responses, such as proliferation, differentiation, or production of cytokines, can be measured and analyzed to gain insights into the immune function or dysfunction.

It's important to note that LCTM is not a routine diagnostic test and its use and interpretation may vary depending on the specific laboratory or research setting.

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.

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.

1. Receptors: In the context of physiology and medicine, receptors are specialized proteins found on the surface of cells or inside cells that detect and respond to specific molecules, known as ligands. These interactions can trigger a range of responses within the cell, such as starting a signaling pathway or changing the cell's behavior. There are various types of receptors, including ion channels, G protein-coupled receptors, and enzyme-linked receptors.

2. Antigen: An antigen is any substance (usually a protein) that can be recognized by the immune system, specifically by antibodies or T-cells, as foreign and potentially harmful. Antigens can be derived from various sources, such as bacteria, viruses, fungi, parasites, or even non-living substances like pollen, chemicals, or toxins. An antigen typically contains epitopes, which are the specific regions that antibodies or T-cell receptors recognize and bind to.

3. T-Cell: Also known as T lymphocytes, T-cells are a type of white blood cell that plays a crucial role in cell-mediated immunity, a part of the adaptive immune system. They are produced in the bone marrow and mature in the thymus gland. There are several types of T-cells, including CD4+ helper T-cells, CD8+ cytotoxic T-cells, and regulatory T-cells (Tregs). T-cells recognize antigens presented to them by antigen-presenting cells (APCs) via their surface receptors called the T-cell receptor (TCR). Once activated, T-cells can proliferate and differentiate into various effector cells that help eliminate infected or damaged cells.

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.

Lymphopenia is a term used in medicine to describe an abnormally low count of lymphocytes, which are a type of white blood cell that plays a crucial role in the body's immune system. Lymphocytes help fight off infections and diseases by producing antibodies and attacking infected cells.

A normal lymphocyte count ranges from 1,000 to 4,800 cells per microliter (cells/μL) of blood in adults. A lymphocyte count lower than 1,000 cells/μL is generally considered lymphopenia.

Several factors can cause lymphopenia, including viral infections, certain medications, autoimmune disorders, and cancer. It's important to note that a low lymphocyte count alone may not indicate a specific medical condition, and further testing may be necessary to determine the underlying cause. If left untreated, lymphopenia can increase the risk of infections and other complications.

Mitogens are substances that stimulate mitosis, or cell division, in particular, the proliferation of cells derived from the immune system. They are often proteins or glycoproteins found on the surface of certain bacteria, viruses, and other cells, which can bind to receptors on the surface of immune cells and trigger a signal transduction pathway that leads to cell division.

Mitogens are commonly used in laboratory research to study the growth and behavior of immune cells, as well as to assess the function of the immune system. For example, mitogens can be added to cultures of lymphocytes (a type of white blood cell) to stimulate their proliferation and measure their response to various stimuli.

Examples of mitogens include phytohemagglutinin (PHA), concanavalin A (ConA), and pokeweed mitogen (PWM). It's important to note that while mitogens can be useful tools in research, they can also have harmful effects if they are introduced into the body in large quantities or inappropriately, as they can stimulate an overactive immune response.

A clone is a group of cells that are genetically identical to each other because they are derived from a common ancestor cell through processes such as mitosis or asexual reproduction. Therefore, the term "clone cells" refers to a population of cells that are genetic copies of a single parent cell.

In the context of laboratory research, cells can be cloned by isolating a single cell and allowing it to divide in culture, creating a population of genetically identical cells. This is useful for studying the behavior and characteristics of individual cell types, as well as for generating large quantities of cells for use in experiments.

It's important to note that while clone cells are genetically identical, they may still exhibit differences in their phenotype (physical traits) due to epigenetic factors or environmental influences.

Lymphocyte depletion is a medical term that refers to the reduction in the number of lymphocytes (a type of white blood cell) in the body. Lymphocytes play a crucial role in the immune system, as they help to fight off infections and diseases.

Lymphocyte depletion can occur due to various reasons, including certain medical treatments such as chemotherapy or radiation therapy, immune disorders, viral infections, or bone marrow transplantation. This reduction in lymphocytes can make a person more susceptible to infections and diseases, as their immune system is weakened.

There are different types of lymphocytes, including T cells, B cells, and natural killer (NK) cells, and lymphocyte depletion can affect one or all of these types. In some cases, lymphocyte depletion may be temporary and resolve on its own or with treatment. However, in other cases, it may be more prolonged and require medical intervention to manage the associated risks and complications.

Immunologic memory, also known as adaptive immunity, refers to the ability of the immune system to recognize and mount a more rapid and effective response upon subsequent exposure to a pathogen or antigen that it has encountered before. This is a key feature of the vertebrate immune system and allows for long-term protection against infectious diseases.

Immunologic memory is mediated by specialized cells called memory T cells and B cells, which are produced during the initial response to an infection or immunization. These cells persist in the body after the pathogen has been cleared and can quickly respond to future encounters with the same or similar antigens. This rapid response leads to a more effective and efficient elimination of the pathogen, resulting in fewer symptoms and reduced severity of disease.

Immunologic memory is the basis for vaccines, which work by exposing the immune system to a harmless form of a pathogen or its components, inducing an initial response and generating memory cells that provide long-term protection against future infections.

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.

Null lymphocytes are a type of immune cells that do not express typical surface markers found on mature T lymphocytes or B lymphocytes. They lack both CD4 and CD8 proteins, which are commonly used to identify T cells, as well as CD19 and CD20 proteins, which are used to identify B cells.

Null lymphocytes can be further divided into two subsets: double negative (DN) and double positive (DP) null cells. DN null cells lack both CD4 and CD8 proteins, while DP null cells express both of these proteins simultaneously. The function of null lymphocytes is not well understood, but they are thought to play a role in the immune response, particularly in the early stages of an infection or inflammation.

It's worth noting that null lymphocytes can also be found in some pathological conditions, such as certain types of leukemia and lymphoma, where they can accumulate in large numbers and contribute to the disease process.

A CD4 lymphocyte count is a laboratory test that measures the number of CD4 T-cells (also known as CD4+ T-cells or helper T-cells) in a sample of blood. CD4 cells are a type of white blood cell that plays a crucial role in the body's immune response, particularly in fighting off infections caused by viruses and other pathogens.

CD4 cells express a protein on their surface called the CD4 receptor, which is used by human immunodeficiency virus (HIV) to infect and destroy these cells. As a result, people with HIV infection or AIDS often have low CD4 lymphocyte counts, which can make them more susceptible to opportunistic infections and other complications.

A normal CD4 lymphocyte count ranges from 500 to 1,200 cells per cubic millimeter of blood (cells/mm3) in healthy adults. A lower than normal CD4 count is often used as a marker for the progression of HIV infection and the development of AIDS. CD4 counts are typically monitored over time to assess the effectiveness of antiretroviral therapy (ART) and to guide clinical decision-making regarding the need for additional interventions, such as prophylaxis against opportunistic infections.

1. Receptors: In the context of physiology and medicine, receptors are specialized proteins found on the surface of cells or inside cells that detect and respond to specific molecules, known as ligands. Receptors play a crucial role in signal transduction, enabling cells to communicate with each other and respond to changes in their environment.
2. Antigen: An antigen is any substance (usually a protein) that can be recognized by the immune system and stimulate an immune response. Antigens can be foreign substances such as bacteria, viruses, or pollen, or they can be components of our own cells, such as tumor antigens in cancer cells. Antigens are typically bound and presented to the immune system by specialized cells called antigen-presenting cells (APCs).
3. T-Cell: T-cells, also known as T lymphocytes, are a type of white blood cell that plays a central role in cell-mediated immunity. T-cells are produced in the bone marrow and mature in the thymus gland. There are two main types of T-cells: CD4+ helper T-cells and CD8+ cytotoxic T-cells. Helper T-cells assist other immune cells, such as B-cells and macrophages, in mounting an immune response, while cytotoxic T-cells directly kill infected or cancerous cells.
4. Alpha-Beta: Alpha-beta is a type of T-cell receptor (TCR) that is found on the surface of most mature T-cells. The alpha-beta TCR is composed of two polypeptide chains, an alpha chain and a beta chain, that are held together by disulfide bonds. The alpha-beta TCR recognizes and binds to specific antigens presented in the context of major histocompatibility complex (MHC) molecules on the surface of APCs. This interaction is critical for initiating an immune response against infected or cancerous cells.

Interleukin-2 (IL-2) receptors are a type of cell surface receptor that bind to and interact with the cytokine interleukin-2. IL-2 is a protein that plays an important role in the immune system, particularly in the activation and proliferation of T cells, a type of white blood cell that helps protect the body from infection and disease.

IL-2 receptors are composed of three subunits: alpha (CD25), beta (CD122), and gamma (CD132). These subunits can combine to form different types of IL-2 receptors, each with different functions. The high-affinity IL-2 receptor is made up of all three subunits and is found on the surface of activated T cells. This type of receptor has a strong binding affinity for IL-2 and plays a crucial role in T cell activation and proliferation.

The intermediate-affinity IL-2 receptor, which consists of the beta and gamma subunits, is found on the surface of resting T cells and natural killer (NK) cells. This type of receptor has a lower binding affinity for IL-2 and plays a role in activating and proliferating these cells.

IL-2 receptors are important targets for immunotherapy, as they play a key role in the regulation of the immune response. Drugs that target IL-2 receptors, such as aldesleukin (Proleukin), have been used to treat certain types of cancer and autoimmune diseases.

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.

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.

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.

An epitope is a specific region on the surface of an antigen (a molecule that can trigger an immune response) that is recognized by an antibody, B-cell receptor, or T-cell receptor. It is also commonly referred to as an antigenic determinant. Epitopes are typically composed of linear amino acid sequences or conformational structures made up of discontinuous amino acids in the antigen. They play a crucial role in the immune system's ability to differentiate between self and non-self molecules, leading to the targeted destruction of foreign substances like viruses and bacteria. Understanding epitopes is essential for developing vaccines, diagnostic tests, and immunotherapies.

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.

Immunoglobulins (Igs), also known as antibodies, are glycoprotein molecules produced by the immune system's B cells in response to the presence of foreign substances, such as bacteria, viruses, and toxins. These Y-shaped proteins play a crucial role in identifying and neutralizing pathogens and other antigens, thereby protecting the body against infection and disease.

Immunoglobulins are composed of four polypeptide chains: two identical heavy chains and two identical light chains, held together by disulfide bonds. The variable regions of these chains form the antigen-binding sites, which recognize and bind to specific epitopes on antigens. Based on their heavy chain type, immunoglobulins are classified into five main isotypes or classes: IgA, IgD, IgE, IgG, and IgM. Each class has distinct functions in the immune response, such as providing protection in different body fluids and tissues, mediating hypersensitivity reactions, and aiding in the development of immunological memory.

In medical settings, immunoglobulins can be administered therapeutically to provide passive immunity against certain diseases or to treat immune deficiencies, autoimmune disorders, and other conditions that may benefit from immunomodulation.

Histocompatibility antigens, also known as human leukocyte antigens (HLAs), are proteins found on the surface of most cells in the body. They play a critical role in the immune system's ability to differentiate between "self" and "non-self" cells. Histocompatibility antigens are encoded by a group of genes called the major histocompatibility complex (MHC).

There are two main types of histocompatibility antigens: class I and class II. Class I antigens are found on almost all nucleated cells, while class II antigens are primarily expressed on immune cells such as B cells, macrophages, and dendritic cells. These antigens present pieces of proteins (peptides) from both inside and outside the cell to T-cells, a type of white blood cell that plays a central role in the immune response.

When foreign peptides are presented to T-cells by histocompatibility antigens, it triggers an immune response aimed at eliminating the threat. This is why histocompatibility antigens are so important in organ transplantation - if the donor's and recipient's antigens do not match closely enough, the recipient's immune system may recognize the transplanted organ as foreign and attack it.

Understanding the role of histocompatibility antigens has been crucial in developing techniques for matching donors and recipients in organ transplantation, as well as in diagnosing and treating various autoimmune diseases and cancers.

CD19 is a type of protein found on the surface of B cells, which are a type of white blood cell that plays a key role in the body's immune response. CD19 is a marker that helps identify and distinguish B cells from other types of cells in the body. It is also a target for immunotherapy in certain diseases, such as B-cell malignancies.

An antigen is any substance that can stimulate an immune response, particularly the production of antibodies. In the context of CD19, antigens refer to substances that can bind to CD19 and trigger a response from the immune system. This can include proteins, carbohydrates, or other molecules found on the surface of bacteria, viruses, or cancer cells.

Therefore, 'antigens, CD19' refers to any substances that can bind to the CD19 protein on B cells and trigger an immune response. These antigens may be used in the development of immunotherapies for the treatment of B-cell malignancies or other diseases.

CD57 is a protein found on the surface of some immune cells, specifically natural killer (NK) cells and certain T-cells. It is often used as a marker to identify these populations of cells. Antigens are substances that can stimulate an immune response, leading to the production of antibodies. In the context of CD57, antigens would refer to any substance that can bind to the CD57 protein on the surface of NK or T-cells.

It's worth noting that CD57 has been studied as a potential marker for certain diseases and conditions, such as HIV infection and some types of cancer. However, its use as a diagnostic or prognostic marker is still a subject of ongoing research and debate.

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.

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.

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.

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).

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.

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.

Jurkat cells are a type of human immortalized T lymphocyte (a type of white blood cell) cell line that is commonly used in scientific research. They were originally isolated from the peripheral blood of a patient with acute T-cell leukemia. Jurkat cells are widely used as a model system to study T-cell activation, signal transduction, and apoptosis (programmed cell death). They are also used in the study of HIV infection and replication, as they can be infected with the virus and used to investigate viral replication and host cell responses.

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.

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.

A lymphocyte transfusion is not a standard medical practice. However, the term "lymphocyte transfusion" generally refers to the infusion of lymphocytes, a type of white blood cell, from a donor to a recipient. This procedure is rarely performed and primarily used in research or experimental settings, such as in the context of adoptive immunotherapy for cancer treatment.

In adoptive immunotherapy, T lymphocytes (a subtype of lymphocytes) are collected from the patient or a donor, activated, expanded in the laboratory, and then reinfused into the patient to enhance their immune response against cancer cells. This is not a common procedure and should only be performed under the guidance of experienced medical professionals in specialized centers.

It's important to note that lymphocyte transfusions are different from stem cell or bone marrow transplants, which involve the infusion of hematopoietic stem cells to reconstitute the recipient's entire blood and immune system.

Lymphocyte cooperation is a term used in immunology to describe the interaction and communication between different types of lymphocytes, specifically T cells and B cells, to mount an effective immune response against pathogens.

T cells, also known as T lymphocytes, are a type of white blood cell that plays a central role in cell-mediated immunity. They can directly kill infected cells or produce cytokines that regulate the immune response. B cells, on the other hand, are responsible for humoral immunity, producing antibodies that neutralize pathogens or mark them for destruction by other immune cells.

Lymphocyte cooperation occurs when a T cell recognizes an antigen presented to it by an antigen-presenting cell (APC) in the context of major histocompatibility complex (MHC) molecules. Once activated, the T cell can then interact with B cells that have also been activated by recognizing the same antigen. The T cell provides help to the B cell by producing cytokines that stimulate its proliferation and differentiation into antibody-secreting plasma cells.

This cooperation between T and B cells is crucial for an effective immune response, as it allows for the generation of a targeted and specific response against pathogens. Defects in lymphocyte cooperation can lead to immunodeficiency or autoimmune disorders.

1. Receptors: In the context of physiology and medicine, receptors are specialized proteins found on the surface of cells or inside cells that detect and respond to specific molecules, known as ligands. These interactions can trigger a variety of responses within the cell, such as starting a signaling cascade or changing the cell's metabolism. Receptors play crucial roles in various biological processes, including communication between cells, regulation of immune responses, and perception of senses.

2. Antigen: An antigen is any substance (usually a protein) that can be recognized by the adaptive immune system, specifically by B-cells and T-cells. Antigens can be derived from various sources, such as microorganisms (like bacteria, viruses, or fungi), pollen, dust mites, or even components of our own cells (for instance, in autoimmune diseases). An antigen's ability to stimulate an immune response is determined by its molecular structure and whether it can be recognized by the receptors on immune cells.

3. B-Cell: B-cells are a type of white blood cell that plays a critical role in the adaptive immune system, particularly in humoral immunity. They originate from hematopoietic stem cells in the bone marrow and are responsible for producing antibodies, which are proteins that recognize and bind to specific antigens. Each B-cell has receptors on its surface called B-cell receptors (BCRs) that can recognize a unique antigen. When a B-cell encounters its specific antigen, it becomes activated, undergoes proliferation, and differentiates into plasma cells that secrete large amounts of antibodies to neutralize or eliminate the antigen.

Pokeweed mitogens are substances derived from the pokeweed plant (Phytolacca americana) that have the ability to stimulate the production and proliferation of various types of cells, particularly white blood cells (lymphocytes). They are often used in laboratory settings as tools for studying the immune system and cell biology.

Pokeweed mitogens are typically extracted from the roots or leaves of the pokeweed plant and purified for use in research and diagnostic applications. When introduced to cells, they bind to specific receptors on the surface of lymphocytes and trigger a series of intracellular signaling events that lead to cell division and growth.

These mitogens are commonly used in immunological assays to measure immune function, such as assessing the proliferative response of lymphocytes to mitogenic stimulation. They can also be used to study the mechanisms of signal transduction and gene regulation in lymphocytes and other cell types.

It is important to note that pokeweed mitogens should only be handled by trained professionals in a controlled laboratory setting, as they can cause adverse reactions if improperly administered or ingested.

CD56 is a type of antigen that is found on the surface of certain cells in the human body. It is also known as neural cell adhesion molecule 1 (NCAM-1) and is a member of the immunoglobulin superfamily. CD56 antigens are primarily expressed on natural killer (NK) cells, a type of immune cell that plays a role in the body's defense against viruses and cancer.

CD56 antigens help NK cells recognize and bind to other cells in the body, such as infected or abnormal cells. This binding can trigger the NK cells to release chemicals that can kill the target cells. CD56 antigens also play a role in the development and function of NK cells, including their ability to communicate with other immune cells and coordinate an effective response to threats.

In addition to NK cells, CD56 antigens are also found on some subsets of T cells, another type of immune cell. In these cells, CD56 antigens help regulate the activation and function of the T cells.

Abnormalities in the expression of CD56 antigens have been associated with various diseases, including certain types of cancer and autoimmune disorders.

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.

CD2 is a type of cell surface protein known as a glycoprotein that is found on the surface of T cells, natural killer (NK) cells, and thymocytes in humans. It plays a role in the activation and regulation of the immune response. CD2 can also function as an adhesion molecule, helping to bind T cells to other cells during an immune response.

An antigen is any substance that can stimulate an immune response, leading to the production of antibodies or the activation of immune cells such as T cells. In the context of CD2, an "antigen" may refer to a specific molecule or structure that interacts with CD2 and triggers a response from T cells or other immune cells.

It's worth noting that while CD2 can interact with certain antigens, it is not itself an antigen in the traditional sense. However, the term "antigen" is sometimes used more broadly to refer to any molecule that interacts with the immune system and triggers a response, so it is possible for CD2 to be referred to as an "antigen" in this context.

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.

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.

CD28 is a co-stimulatory molecule that plays an important role in the activation and regulation of T cells, which are key players in the immune response. It is a type of protein found on the surface of T cells and interacts with other proteins called B7-1 (also known as CD80) and B7-2 (also known as CD86) that are expressed on the surface of antigen-presenting cells (APCs).

When a T cell encounters an APC that is presenting an antigen, the T cell receptor (TCR) on the surface of the T cell recognizes and binds to the antigen. However, this interaction alone is not enough to fully activate the T cell. The engagement of CD28 with B7-1 or B7-2 provides a critical co-stimulatory signal that promotes T cell activation, proliferation, and survival.

CD28 is also an important target for immune checkpoint inhibitors, which are drugs used to treat cancer by blocking the inhibitory signals that prevent T cells from attacking tumor cells. By blocking CD28, these drugs can enhance the anti-tumor response of T cells and improve cancer outcomes.

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.

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.

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.

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.

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.

Immunoglobulin M (IgM) is a type of antibody that is primarily found in the blood and lymph fluid. It is the first antibody to be produced in response to an initial exposure to an antigen, making it an important part of the body's primary immune response. IgM antibodies are large molecules that are composed of five basic units, giving them a pentameric structure. They are primarily found on the surface of B cells as membrane-bound immunoglobulins (mlgM), where they function as receptors for antigens. Once an mlgM receptor binds to an antigen, it triggers the activation and differentiation of the B cell into a plasma cell that produces and secretes large amounts of soluble IgM antibodies.

IgM antibodies are particularly effective at agglutination (clumping) and complement activation, which makes them important in the early stages of an immune response to help clear pathogens from the bloodstream. However, they are not as stable or long-lived as other types of antibodies, such as IgG, and their levels tend to decline after the initial immune response has occurred.

In summary, Immunoglobulin M (IgM) is a type of antibody that plays a crucial role in the primary immune response to antigens by agglutination and complement activation. It is primarily found in the blood and lymph fluid, and it is produced by B cells after they are activated by an antigen.

The palatine tonsils, also known as the "tonsils," are two masses of lymphoid tissue located on either side of the oropharynx, at the back of the throat. They are part of the immune system and play a role in protecting the body from inhaled or ingested pathogens. Each tonsil has a surface covered with crypts and follicles that contain lymphocytes, which help to filter out bacteria and viruses that enter the mouth and nose.

The palatine tonsils are visible through the mouth and can be seen during a routine physical examination. They vary in size, but typically are about the size of a large olive or almond. Swelling or inflammation of the tonsils is called tonsillitis, which can cause symptoms such as sore throat, difficulty swallowing, fever, and swollen lymph nodes in the neck. In some cases, enlarged tonsils may need to be removed through a surgical procedure called a tonsillectomy.

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.

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 "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.

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.

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).

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.

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.

Lymphocyte Function-Associated Antigen-1 (LFA-1) is a type of integrin, which is a family of cell surface proteins that are important for cell-cell adhesion and signal transduction. LFA-1 is composed of two subunits, called alpha-L (CD11a) and beta-2 (CD18), and it is widely expressed on various leukocytes, including T cells, B cells, and natural killer cells.

LFA-1 plays a crucial role in the immune system by mediating the adhesion of leukocytes to other cells, such as endothelial cells that line blood vessels, and extracellular matrix components. This adhesion is necessary for leukocyte migration from the bloodstream into tissues during inflammation or immune responses. LFA-1 also contributes to the activation of T cells and their interaction with antigen-presenting cells, such as dendritic cells and macrophages.

The binding of LFA-1 to its ligands, including intercellular adhesion molecule 1 (ICAM-1) and ICAM-2, triggers intracellular signaling pathways that regulate various cellular functions, such as cytoskeletal reorganization, gene expression, and cell survival. Dysregulation of LFA-1 function has been implicated in several immune-related diseases, including autoimmune disorders, inflammatory diseases, and cancer.

L-Selectin, also known as LECAM-1 (Leukocyte Cell Adhesion Molecule 1), is a type of cell adhesion molecule that is found on the surface of leukocytes (white blood cells). It plays an important role in the immune system by mediating the initial attachment and rolling of leukocytes along the endothelial lining of blood vessels, which is a critical step in the process of inflammation and immune response.

L-Selectin recognizes specific sugar structures called sialyl Lewis x (sLeX) and related structures on the surface of endothelial cells, allowing leukocytes to bind to them. This interaction helps to slow down the leukocytes and facilitate their extravasation from the blood vessels into the surrounding tissues, where they can carry out their immune functions.

L-Selectin is involved in a variety of immunological processes, including the recruitment of leukocytes to sites of infection or injury, the homing of lymphocytes to lymphoid organs, and the regulation of immune cell trafficking under homeostatic conditions.

Lymphocyte homing receptors are specialized molecules found on the surface of lymphocytes (white blood cells that include T-cells and B-cells), which play a crucial role in the immune system's response to infection and disease. These receptors facilitate the targeted migration and trafficking of lymphocytes from the bloodstream to specific secondary lymphoid organs, such as lymph nodes, spleen, and Peyer's patches in the intestines, where they can encounter antigens and mount an immune response.

The homing receptors consist of two main components: adhesion molecules and chemokine receptors. Adhesion molecules, such as selectins and integrins, mediate the initial attachment and rolling of lymphocytes along the endothelial cells that line the blood vessels in lymphoid organs. Chemokine receptors, on the other hand, interact with chemokines (a type of cytokine) that are secreted by the endothelial cells and stromal cells within the lymphoid organs. This interaction triggers a signaling cascade that activates integrins, leading to their firm adhesion to the endothelium and subsequent transmigration into the lymphoid tissue.

The specificity of this homing process is determined by the unique combination of adhesion molecules and chemokine receptors expressed on different subsets of lymphocytes, which allows them to home to distinct anatomical locations in response to various chemokine gradients. This targeted migration ensures that the immune system can effectively mount a rapid and localized response against pathogens while minimizing unnecessary inflammation in other parts of the body.

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.

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.

Immunologic deficiency syndromes refer to a group of disorders characterized by defective functioning of the immune system, leading to increased susceptibility to infections and malignancies. These deficiencies can be primary (genetic or congenital) or secondary (acquired due to environmental factors, medications, or diseases).

Primary immunodeficiency syndromes (PIDS) are caused by inherited genetic mutations that affect the development and function of immune cells, such as T cells, B cells, and phagocytes. Examples include severe combined immunodeficiency (SCID), common variable immunodeficiency (CVID), Wiskott-Aldrich syndrome, and X-linked agammaglobulinemia.

Secondary immunodeficiency syndromes can result from various factors, including:

1. HIV/AIDS: Human Immunodeficiency Virus infection leads to the depletion of CD4+ T cells, causing profound immune dysfunction and increased vulnerability to opportunistic infections and malignancies.
2. Medications: Certain medications, such as chemotherapy, immunosuppressive drugs, and long-term corticosteroid use, can impair immune function and increase infection risk.
3. Malnutrition: Deficiencies in essential nutrients like protein, vitamins, and minerals can weaken the immune system and make individuals more susceptible to infections.
4. Aging: The immune system naturally declines with age, leading to an increased incidence of infections and poorer vaccine responses in older adults.
5. Other medical conditions: Chronic diseases such as diabetes, cancer, and chronic kidney or liver disease can also compromise the immune system and contribute to immunodeficiency syndromes.

Immunologic deficiency syndromes require appropriate diagnosis and management strategies, which may include antimicrobial therapy, immunoglobulin replacement, hematopoietic stem cell transplantation, or targeted treatments for the underlying cause.

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.

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.

'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.

An epitope is a specific region on an antigen (a substance that triggers an immune response) that is recognized and bound by an antibody or a T-cell receptor. In the case of T-lymphocytes, which are a type of white blood cell that plays a central role in cell-mediated immunity, epitopes are typically presented on the surface of infected cells in association with major histocompatibility complex (MHC) molecules.

T-lymphocytes recognize and respond to epitopes through their T-cell receptors (TCRs), which are membrane-bound proteins that can bind to specific epitopes presented on the surface of infected cells. There are two main types of T-lymphocytes: CD4+ T-cells, also known as helper T-cells, and CD8+ T-cells, also known as cytotoxic T-cells.

CD4+ T-cells recognize epitopes presented in the context of MHC class II molecules, which are typically expressed on the surface of professional antigen-presenting cells such as dendritic cells, macrophages, and B-cells. CD4+ T-cells help to coordinate the immune response by producing cytokines that activate other immune cells.

CD8+ T-cells recognize epitopes presented in the context of MHC class I molecules, which are expressed on the surface of almost all nucleated cells. CD8+ T-cells are able to directly kill infected cells by releasing cytotoxic granules that contain enzymes that can induce apoptosis (programmed cell death) in the target cell.

In summary, epitopes are specific regions on antigens that are recognized and bound by T-lymphocytes through their T-cell receptors. CD4+ T-cells recognize epitopes presented in the context of MHC class II molecules, while CD8+ T-cells recognize epitopes presented in the context of MHC class I molecules.

"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.

Reference values, also known as reference ranges or reference intervals, are the set of values that are considered normal or typical for a particular population or group of people. These values are often used in laboratory tests to help interpret test results and determine whether a patient's value falls within the expected range.

The process of establishing reference values typically involves measuring a particular biomarker or parameter in a large, healthy population and then calculating the mean and standard deviation of the measurements. Based on these statistics, a range is established that includes a certain percentage of the population (often 95%) and excludes extreme outliers.

It's important to note that reference values can vary depending on factors such as age, sex, race, and other demographic characteristics. Therefore, it's essential to use reference values that are specific to the relevant population when interpreting laboratory test results. Additionally, reference values may change over time due to advances in measurement technology or changes in the population being studied.

Feline Acquired Immunodeficiency Syndrome (FAIDS) is a progressive immune disorder in cats caused by infection with the feline immunodeficiency virus (FIV). The virus attacks and weakens the cat's immune system, making it difficult for the animal to fight off other infections and diseases.

The initial infection with FIV may cause symptoms such as fever, swollen lymph nodes, and loss of appetite. However, many cats do not show any signs of illness for years after the initial infection. As the immune system becomes weaker over time, the cat becomes more susceptible to various secondary infections, cancers, and other diseases. Common symptoms in advanced stages of FAIDS include weight loss, chronic or recurring infections (such as respiratory, skin, or gastrointestinal infections), dental disease, anemia, and neurological disorders.

FAIDS is most commonly spread through bite wounds from infected cats, as the virus is present in their saliva. It can also be transmitted through sexual contact or from mother to kitten during pregnancy or nursing. There is no cure for FAIDS, but antiretroviral therapy (ART) can help manage the infection and slow down its progression. Supportive care, such as proper nutrition, regular veterinary check-ups, and monitoring for secondary infections, is essential for maintaining the cat's quality of life.

It is important to note that FIV is species-specific and cannot be transmitted from cats to humans or other animals, except non-human primates.

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.

I'm happy to help! "Rats, Inbred Lew" is a specific strain of laboratory rats that have been inbred for research purposes. The "Lew" part of the name refers to the location where they were first developed, the Lewis Institute in Lake Bluff, Illinois, USA.

Inbreeding is a process of mating closely related individuals over many generations to create a genetically homogeneous population. This results in a high degree of genetic similarity among members of the strain, making them ideal for use as experimental models because any differences observed between individuals are more likely to be due to the experimental manipulation rather than genetic variation.

Inbred Lew rats have been widely used in biomedical research, particularly in studies related to hypertension and cardiovascular disease. They exhibit a number of unique characteristics that make them useful for these types of studies, including their susceptibility to developing high blood pressure when fed a high-salt diet or given certain drugs.

It's important to note that while inbred strains like Lew rats can be very useful tools for researchers, they are not perfect models for human disease. Because they have been bred in a controlled environment and selected for specific traits, they may not respond to experimental manipulations in the same way that humans or other animals would. Therefore, it's important to interpret findings from these studies with caution and consider multiple lines of evidence before drawing any firm conclusions.

Neoplasm antigens, also known as tumor antigens, are substances that are produced by cancer cells (neoplasms) and can stimulate an immune response. These antigens can be proteins, carbohydrates, or other molecules that are either unique to the cancer cells or are overexpressed or mutated versions of normal cellular proteins.

Neoplasm antigens can be classified into two main categories: tumor-specific antigens (TSAs) and tumor-associated antigens (TAAs). TSAs are unique to cancer cells and are not expressed by normal cells, while TAAs are present at low levels in normal cells but are overexpressed or altered in cancer cells.

TSAs can be further divided into viral antigens and mutated antigens. Viral antigens are produced when cancer is caused by a virus, such as human papillomavirus (HPV) in cervical cancer. Mutated antigens are the result of genetic mutations that occur during cancer development and are unique to each patient's tumor.

Neoplasm antigens play an important role in the immune response against cancer. They can be recognized by the immune system, leading to the activation of immune cells such as T cells and natural killer (NK) cells, which can then attack and destroy cancer cells. However, cancer cells often develop mechanisms to evade the immune response, allowing them to continue growing and spreading.

Understanding neoplasm antigens is important for the development of cancer immunotherapies, which aim to enhance the body's natural immune response against cancer. These therapies include checkpoint inhibitors, which block proteins that inhibit T cell activation, and therapeutic vaccines, which stimulate an immune response against specific tumor antigens.

HLA-A2 antigen is a type of human leukocyte antigen (HLA) class I molecule, which is found on the surface of cells in our body. HLA molecules are responsible for presenting pieces of proteins (peptides) from inside the cell to the immune system's T-cells, helping them distinguish between "self" and "non-self" proteins.

HLA-A2 is one of the most common HLA class I antigens in the Caucasian population, with an estimated frequency of around 50%. It presents a variety of peptides to T-cells, including those derived from viruses and tumor cells. The presentation of these peptides can trigger an immune response, leading to the destruction of infected or malignant cells.

It is important to note that HLA typing is crucial in organ transplantation, as a mismatch between donor and recipient HLA antigens can lead to rejection of the transplanted organ. Additionally, HLA-A2 has been associated with certain autoimmune diseases and cancer types, making it an area of interest for researchers studying these conditions.

Lymphocytosis is a medical term that refers to an abnormal increase in the number of lymphocytes (a type of white blood cell) in the peripheral blood. A normal lymphocyte count ranges from 1,000 to 4,800 cells per microliter (μL) of blood in adults. Lymphocytosis is typically defined as a lymphocyte count greater than 4,800 cells/μL in adults or higher than age-specific normal values in children.

There are various causes of lymphocytosis, including viral infections (such as mononucleosis), bacterial infections, tuberculosis, fungal infections, parasitic infections, autoimmune disorders, allergies, and certain cancers like chronic lymphocytic leukemia or lymphoma. It is essential to investigate the underlying cause of lymphocytosis through a thorough clinical evaluation, medical history, physical examination, and appropriate diagnostic tests, such as blood tests, imaging studies, or biopsies.

It's important to note that an isolated episode of mild lymphocytosis is often not clinically significant and may resolve on its own without any specific treatment. However, persistent or severe lymphocytosis requires further evaluation and management based on the underlying cause.

Immunocompetence is the condition of having a properly functioning immune system that can effectively respond to the presence of foreign substances, such as pathogens (like bacteria, viruses, and parasites) and other potentially harmful agents. It involves the ability of the immune system to recognize, attack, and eliminate these foreign substances while also maintaining tolerance to self-tissues and promoting tissue repair.

Immunocompetence is essential for overall health and wellbeing, as it helps protect the body from infections and diseases. Factors that can affect immunocompetence include age, genetics, stress, nutrition, sleep, and certain medical conditions or treatments (like chemotherapy or immunosuppressive drugs) that can weaken the immune system.

CD5 is a type of protein found on the surface of certain cells in the human body, including some immune cells like T cells and B cells. It is also known as a cell marker or identifier. Antigens are substances (usually proteins) on the surface of cells that can be recognized by the immune system, triggering an immune response.

In the context of CD5, antigens refer to foreign substances that can bind to the CD5 protein and stimulate an immune response. However, it's important to note that CD5 itself is not typically considered an antigen in the medical community. Instead, it is a marker used to identify certain types of cells and monitor their behavior in health and disease states.

In some cases, abnormal expression or regulation of CD5 has been associated with various diseases, including certain types of cancer. For example, some B-cell lymphomas may overexpress CD5, which can help doctors diagnose and monitor the progression of the disease. However, in these contexts, CD5 is not considered an antigen in the traditional sense.

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.

Lymph is a colorless, transparent fluid that circulates throughout the lymphatic system, which is a part of the immune and circulatory systems. It consists of white blood cells called lymphocytes, proteins, lipids, glucose, electrolytes, hormones, and waste products. Lymph plays an essential role in maintaining fluid balance, absorbing fats from the digestive tract, and defending the body against infection by transporting immune cells to various tissues and organs. It is collected from tissues through lymph capillaries and flows through increasingly larger lymphatic vessels, ultimately returning to the bloodstream via the subclavian veins in the chest region.

Autoimmune diseases are a group of disorders in which the immune system, which normally protects the body from foreign invaders like bacteria and viruses, mistakenly attacks the body's own cells and tissues. This results in inflammation and damage to various organs and tissues in the body.

In autoimmune diseases, the body produces autoantibodies that target its own proteins or cell receptors, leading to their destruction or malfunction. The exact cause of autoimmune diseases is not fully understood, but it is believed that a combination of genetic and environmental factors contribute to their development.

There are over 80 different types of autoimmune diseases, including rheumatoid arthritis, lupus, multiple sclerosis, type 1 diabetes, Hashimoto's thyroiditis, Graves' disease, psoriasis, and inflammatory bowel disease. Symptoms can vary widely depending on the specific autoimmune disease and the organs or tissues affected. Treatment typically involves managing symptoms and suppressing the immune system to prevent further damage.

Blood cells are the formed elements in the blood, including red blood cells (erythrocytes), white blood cells (leukocytes), and platelets (thrombocytes). These cells are produced in the bone marrow and play crucial roles in the body's functions. Red blood cells are responsible for carrying oxygen to tissues and carbon dioxide away from them, while white blood cells are part of the immune system and help defend against infection and disease. Platelets are cell fragments that are essential for normal blood clotting.

'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.

Adoptive transfer is a medical procedure in which immune cells are transferred from a donor to a recipient with the aim of providing immunity or treating a disease, such as cancer. This technique is often used in the field of immunotherapy and involves isolating specific immune cells (like T-cells) from the donor, expanding their numbers in the laboratory, and then infusing them into the patient. The transferred cells are expected to recognize and attack the target cells, such as malignant or infected cells, leading to a therapeutic effect. This process requires careful matching of donor and recipient to minimize the risk of rejection and graft-versus-host disease.

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.

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.

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.

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.

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)

Perforin is a protein that plays a crucial role in the immune system's response to virally infected or cancerous cells. It is primarily produced and released by cytotoxic T-cells and natural killer (NK) cells, two types of white blood cells involved in defending the body against infection and disease.

Perforin functions by creating pores or holes in the membrane of target cells, leading to their lysis or destruction. This process allows for the release of cellular contents and the exposure of intracellular antigens, which can then be processed and presented to other immune cells, thereby enhancing the immune response against the pathogen or abnormal cells.

In summary, perforin is a vital component of the immune system's cytotoxic activity, contributing to the elimination of infected or malignant cells and maintaining overall health and homeostasis in the body.

The appendix is a small, tube-like structure that projects from the large intestine, located in the lower right quadrant of the abdomen. Its function in humans is not well understood and is often considered vestigial, meaning it no longer serves a necessary purpose. However, in some animals, the appendix plays a role in the immune system. Inflammation of the appendix, known as appendicitis, can cause severe abdominal pain and requires medical attention, often leading to surgical removal of the appendix (appendectomy).

Immunotherapy is a type of medical treatment that uses the body's own immune system to fight against diseases, such as cancer. It involves the use of substances (like vaccines, medications, or immune cells) that stimulate or suppress the immune system to help it recognize and destroy harmful disease-causing cells or agents, like tumor cells.

Immunotherapy can work in several ways:

1. Activating the immune system: Certain immunotherapies boost the body's natural immune responses, helping them recognize and attack cancer cells more effectively.
2. Suppressing immune system inhibitors: Some immunotherapies target and block proteins or molecules that can suppress the immune response, allowing the immune system to work more efficiently against diseases.
3. Replacing or enhancing specific immune cells: Immunotherapy can also involve administering immune cells (like T-cells) that have been genetically engineered or modified to recognize and destroy cancer cells.

Immunotherapies have shown promising results in treating various types of cancer, autoimmune diseases, and allergies. However, they can also cause side effects, as an overactive immune system may attack healthy tissues and organs. Therefore, careful monitoring is necessary during immunotherapy treatment.

Homologous transplantation is a type of transplant surgery where organs or tissues are transferred between two genetically non-identical individuals of the same species. The term "homologous" refers to the similarity in structure and function of the donated organ or tissue to the recipient's own organ or tissue.

For example, a heart transplant from one human to another is an example of homologous transplantation because both organs are hearts and perform the same function. Similarly, a liver transplant, kidney transplant, lung transplant, and other types of organ transplants between individuals of the same species are also considered homologous transplantations.

Homologous transplantation is in contrast to heterologous or xenogeneic transplantation, where organs or tissues are transferred from one species to another, such as a pig heart transplanted into a human. Homologous transplantation is more commonly performed than heterologous transplantation due to the increased risk of rejection and other complications associated with xenogeneic transplants.

Antilymphocyte serum (ALS) is a type of immune serum that contains antibodies against human lymphocytes. It is produced by immunizing animals, such as horses or rabbits, with human lymphocytes to stimulate an immune response and the production of anti-lymphocyte antibodies. The resulting serum is then collected and can be used as a therapeutic agent to suppress the activity of the immune system in certain medical conditions.

ALS is primarily used in the treatment of transplant rejection, particularly in organ transplantation, where it helps to prevent the recipient's immune system from attacking and rejecting the transplanted organ. It can also be used in the management of autoimmune diseases, such as rheumatoid arthritis and lupus, to suppress the overactive immune response that contributes to these conditions.

It is important to note that the use of ALS carries a risk of side effects, including allergic reactions, fever, and decreased white blood cell counts. Close monitoring and appropriate management of these potential adverse events are essential during treatment with ALS.

H-2 antigens are a group of cell surface proteins found in mice that play a critical role in the immune system. They are similar to the human leukocyte antigen (HLA) complex in humans and are involved in the presentation of peptide antigens to T cells, which is a crucial step in the adaptive immune response.

The H-2 antigens are encoded by a cluster of genes located on chromosome 17 in mice. They are highly polymorphic, meaning that there are many different variations of these proteins circulating in the population. This genetic diversity allows for a wide range of potential peptide antigens to be presented to T cells, thereby enhancing the ability of the immune system to recognize and respond to a variety of pathogens.

The H-2 antigens are divided into two classes based on their function and structure. Class I H-2 antigens are found on almost all nucleated cells and consist of a heavy chain, a light chain, and a peptide fragment. They present endogenous peptides, such as those derived from viruses that infect the cell, to CD8+ T cells.

Class II H-2 antigens, on the other hand, are found primarily on professional antigen-presenting cells, such as dendritic cells and macrophages. They consist of an alpha chain and a beta chain and present exogenous peptides, such as those derived from bacteria that have been engulfed by the cell, to CD4+ T cells.

Overall, H-2 antigens are essential components of the mouse immune system, allowing for the recognition and elimination of pathogens and infected cells.

Histocompatibility antigens, class I are proteins found on the surface of most cells in the body. They play a critical role in the immune system's ability to differentiate between "self" and "non-self." These antigens are composed of three polypeptides - two heavy chains and one light chain - and are encoded by genes in the major histocompatibility complex (MHC) on chromosome 6 in humans.

Class I MHC molecules present peptide fragments from inside the cell to CD8+ T cells, also known as cytotoxic T cells. This presentation allows the immune system to detect and destroy cells that have been infected by viruses or other intracellular pathogens, or that have become cancerous.

There are three main types of class I MHC molecules in humans: HLA-A, HLA-B, and HLA-C. The term "HLA" stands for human leukocyte antigen, which reflects the original identification of these proteins on white blood cells (leukocytes). The genes encoding these molecules are highly polymorphic, meaning there are many different variants in the population, and matching HLA types is essential for successful organ transplantation to minimize the risk of rejection.

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.

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.

Immunosuppressive agents are medications that decrease the activity of the immune system. They are often used to prevent the rejection of transplanted organs and to treat autoimmune diseases, where the immune system mistakenly attacks the body's own tissues. These drugs work by interfering with the immune system's normal responses, which helps to reduce inflammation and damage to tissues. However, because they suppress the immune system, people who take immunosuppressive agents are at increased risk for infections and other complications. Examples of immunosuppressive agents include corticosteroids, azathioprine, cyclophosphamide, mycophenolate mofetil, tacrolimus, and sirolimus.

Cyclosporins are a group of cyclic undecapeptides that have immunosuppressive properties. The most well-known and widely used cyclosporin is cyclosporine A, which is commonly used in organ transplantation to prevent rejection. It works by inhibiting the activation of T-cells, a type of white blood cell that plays a central role in the immune response. By suppressing the activity of T-cells, cyclosporine A reduces the risk of an immune response against the transplanted organ.

Cyclosporins are also used in the treatment of autoimmune diseases, such as rheumatoid arthritis and psoriasis, where they help to reduce inflammation and prevent damage to tissues. Like all immunosuppressive drugs, cyclosporins can increase the risk of infection and cancer, so they must be used with caution and under close medical supervision.

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.

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.

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.

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.

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.

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.

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.

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.

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.

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.

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.

Acquired Immunodeficiency Syndrome (AIDS) is a chronic, life-threatening condition caused by the Human Immunodeficiency Virus (HIV). AIDS is the most advanced stage of HIV infection, characterized by the significant weakening of the immune system, making the person more susceptible to various opportunistic infections and cancers.

The medical definition of AIDS includes specific criteria based on CD4+ T-cell count or the presence of certain opportunistic infections and diseases. According to the Centers for Disease Control and Prevention (CDC), a person with HIV is diagnosed with AIDS when:

1. The CD4+ T-cell count falls below 200 cells per cubic millimeter of blood (mm3) - a normal range is typically between 500 and 1,600 cells/mm3.
2. They develop one or more opportunistic infections or cancers that are indicative of advanced HIV disease, regardless of their CD4+ T-cell count.

Some examples of these opportunistic infections and cancers include:

* Pneumocystis pneumonia (PCP)
* Candidiasis (thrush) affecting the esophagus, trachea, or lungs
* Cryptococcal meningitis
* Toxoplasmosis of the brain
* Cytomegalovirus disease
* Kaposi's sarcoma
* Non-Hodgkin's lymphoma
* Invasive cervical cancer

It is important to note that with appropriate antiretroviral therapy (ART), people living with HIV can maintain their CD4+ T-cell counts, suppress viral replication, and prevent the progression to AIDS. Early diagnosis and consistent treatment are crucial for managing HIV and improving life expectancy and quality of life.

Adoptive immunotherapy is a type of cancer treatment that involves the removal of immune cells from a patient, followed by their modification and expansion in the laboratory, and then reinfusion back into the patient to help boost their immune system's ability to fight cancer. This approach can be used to enhance the natural ability of T-cells (a type of white blood cell) to recognize and destroy cancer cells.

There are different types of adoptive immunotherapy, including:

1. T-cell transfer therapy: In this approach, T-cells are removed from the patient's tumor or blood, activated and expanded in the laboratory, and then reinfused back into the patient. Some forms of T-cell transfer therapy involve genetically modifying the T-cells to express chimeric antigen receptors (CARs) that recognize specific proteins on the surface of cancer cells.
2. Tumor-infiltrating lymphocyte (TIL) therapy: This type of adoptive immunotherapy involves removing T-cells directly from a patient's tumor, expanding them in the laboratory, and then reinfusing them back into the patient. The expanded T-cells are specifically targeted to recognize and destroy cancer cells.
3. Dendritic cell (DC) vaccine: DCs are specialized immune cells that help activate T-cells. In this approach, DCs are removed from the patient, exposed to tumor antigens in the laboratory, and then reinfused back into the patient to stimulate a stronger immune response against cancer cells.

Adoptive immunotherapy has shown promise in treating certain types of cancer, such as melanoma and leukemia, but more research is needed to determine its safety and efficacy in other types of cancer.

The term "Immune Adherence Reaction" is not widely used in modern immunology or medicine. It appears to be an outdated concept that refers to the attachment of immune complexes (consisting of antigens, antibodies, and complement components) to Fc receptors on phagocytic cells, such as neutrophils and monocytes. This interaction facilitates the clearance of immune complexes from circulation and helps to prevent tissue damage caused by their deposition.

However, it is important to note that this term is not commonly used in current scientific literature or clinical settings. Instead, the processes it describes are typically discussed within the broader context of immune complex-mediated inflammation, complement activation, and phagocytosis.

Pore-forming cytotoxic proteins are a group of toxins that can create pores or holes in the membranes of cells, leading to cell damage or death. These toxins are produced by various organisms, including bacteria, fungi, and plants, as a defense mechanism or to help establish an infection.

The pore-forming cytotoxic proteins can be divided into two main categories:

1. Membrane attack complex/perforin (MACPF) domain-containing proteins: These are found in many organisms, including humans. They form pores by oligomerizing, or clustering together, in the target cell membrane. An example of this type of toxin is the perforin protein, which is released by cytotoxic T cells and natural killer cells to destroy virus-infected or cancerous cells.
2. Cholesterol-dependent cytolysins (CDCs): These are mainly produced by gram-positive bacteria. They bind to cholesterol in the target cell membrane, forming a prepore structure that then undergoes conformational changes to create a pore. An example of a CDC is alpha-hemolysin from Staphylococcus aureus, which can lyse red blood cells and damage various other cell types.

These pore-forming cytotoxic proteins play a significant role in host-pathogen interactions and have implications for the development of novel therapeutic strategies.

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.

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.

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.

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.

Granzymes are a group of proteases (enzymes that break down other proteins) that are stored in the granules of cytotoxic T cells and natural killer (NK) cells. They play an important role in the immune response by inducing apoptosis (programmed cell death) in target cells, such as virus-infected or cancer cells. Granzymes are released into the immunological synapse between the effector and target cells, where they can enter the target cell and cleave specific substrates, leading to the activation of caspases and ultimately apoptosis. There are several different types of granzymes, each with distinct substrate specificities and functions.

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.

Melanoma is defined as a type of cancer that develops from the pigment-containing cells known as melanocytes. It typically occurs in the skin but can rarely occur in other parts of the body, including the eyes and internal organs. Melanoma is characterized by the uncontrolled growth and multiplication of melanocytes, which can form malignant tumors that invade and destroy surrounding tissue.

Melanoma is often caused by exposure to ultraviolet (UV) radiation from the sun or tanning beds, but it can also occur in areas of the body not exposed to the sun. It is more likely to develop in people with fair skin, light hair, and blue or green eyes, but it can affect anyone, regardless of their skin type.

Melanoma can be treated effectively if detected early, but if left untreated, it can spread to other parts of the body and become life-threatening. Treatment options for melanoma include surgery, radiation therapy, chemotherapy, immunotherapy, and targeted therapy, depending on the stage and location of the cancer. Regular skin examinations and self-checks are recommended to detect any changes or abnormalities in moles or other pigmented lesions that may indicate melanoma.

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.

'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.

CD27 is a protein that is found on the surface of certain immune cells, including T cells and B cells. It is a type of molecule known as a cell-surface antigen, which can be recognized by other immune cells and used to target those cells for activation or destruction. CD27 plays a role in the regulation of the immune response, particularly in the activation and differentiation of T cells.

CD27 is also a member of the tumor necrosis factor receptor (TNFR) superfamily, which means that it has a specific structure and function that allows it to interact with other molecules called ligands. The interaction between CD27 and its ligand, CD70, helps to activate T cells and promote their survival and proliferation.

In addition to its role in the immune response, CD27 has also been studied as a potential target for cancer immunotherapy. Because CD27 is expressed on certain types of tumor cells, it may be possible to use therapies that target CD27 to stimulate an immune response against the tumor and help to destroy it. However, more research is needed to determine the safety and effectiveness of these approaches.

Leukemia, lymphoid is a type of cancer that affects the lymphoid cells, which are a vital part of the body's immune system. It is characterized by the uncontrolled production of abnormal white blood cells (leukocytes or WBCs) in the bone marrow, specifically the lymphocytes. These abnormal lymphocytes accumulate and interfere with the production of normal blood cells, leading to a decrease in red blood cells (anemia), platelets (thrombocytopenia), and healthy white blood cells (leukopenia).

There are two main types of lymphoid leukemia: acute lymphoblastic leukemia (ALL) and chronic lymphocytic leukemia (CLL). Acute lymphoblastic leukemia progresses rapidly, while chronic lymphocytic leukemia has a slower onset and progression.

Symptoms of lymphoid leukemia may include fatigue, frequent infections, easy bruising or bleeding, weight loss, swollen lymph nodes, and bone pain. Treatment options depend on the type, stage, and individual patient factors but often involve chemotherapy, radiation therapy, targeted therapy, immunotherapy, or stem cell transplantation.

CD95 (also known as Fas or APO-1) is a type of cell surface receptor that can bind to specific proteins and trigger programmed cell death, also known as apoptosis. It is an important regulator of the immune system and helps to control the activation and deletion of immune cells. CD95 ligand (CD95L), the protein that binds to CD95, is expressed on activated T-cells and can induce apoptosis in other cells that express CD95, including other T-cells and tumor cells.

An antigen is any substance that can stimulate an immune response, leading to the production of antibodies or activation of immune cells. In the context of CD95, antigens may refer to substances that can induce the expression of CD95 on the surface of cells, making them susceptible to CD95L-mediated apoptosis. These antigens could include viral proteins, tumor antigens, or other substances that trigger an immune response.

Therefore, the medical definition of 'antigens, CD95' may refer to substances that can induce the expression of CD95 on the surface of cells and make them targets for CD95L-mediated apoptosis.

Feline Immunodeficiency Virus (FIV) is a lentivirus that primarily affects felines, including domestic cats and wild cats. It is the feline equivalent of Human Immunodeficiency Virus (HIV). The virus attacks the immune system, specifically the CD4+ T-cells, leading to a decline in the immune function over time.

This makes the infected cat more susceptible to various secondary infections and diseases. It is usually transmitted through bite wounds from infected cats during fighting or mating. Mother to offspring transmission can also occur, either in utero, during birth, or through nursing.

There is no cure for FIV, but antiretroviral therapy can help manage the disease and improve the quality of life for infected cats. It's important to note that while FIV-positive cats can live normal lives for many years, they should be kept indoors to prevent transmission to other cats and to protect them from opportunistic infections.

Fas Ligand Protein (FasL or CD95L) is a type II transmembrane protein belonging to the tumor necrosis factor (TNF) superfamily. It plays a crucial role in programmed cell death, also known as apoptosis. The FasL protein binds to its receptor, Fas (CD95 or APO-1), which is found on the surface of various cells including immune cells. This binding triggers a signaling cascade that leads to apoptosis, helping to regulate the immune response and maintain homeostasis in tissues.

FasL can also be produced as a soluble protein (sFasL) through alternative splicing or proteolytic cleavage of the membrane-bound form. Soluble FasL may have different functions compared to its membrane-bound counterpart, and its role in physiology and disease is still under investigation.

Dysregulation of the Fas/FasL system has been implicated in various pathological conditions, including autoimmune diseases, neurodegenerative disorders, and cancer.

The Interleukin-2 Receptor alpha Subunit (IL-2Rα), also known as CD25, is a protein that is expressed on the surface of certain immune cells, such as activated T-cells and B-cells. It is a subunit of the interleukin-2 receptor, which plays a crucial role in the activation and regulation of the immune response. The IL-2Rα binds to interleukin-2 (IL-2) with high affinity, forming a complex that initiates intracellular signaling pathways involved in T-cell proliferation, differentiation, and survival. IL-2Rα is also a target for immunosuppressive therapies used to prevent rejection of transplanted organs and to treat autoimmune diseases.

CD20 is not a medical definition of an antigen, but rather it is a cell surface marker that helps identify a specific type of white blood cell called B-lymphocytes or B-cells. These cells are part of the adaptive immune system and play a crucial role in producing antibodies to fight off infections.

CD20 is a protein found on the surface of mature B-cells, and it is used as a target for monoclonal antibody therapies in the treatment of certain types of cancer and autoimmune diseases. Rituximab is an example of a monoclonal antibody that targets CD20 and is used to treat conditions such as non-Hodgkin lymphoma, chronic lymphocytic leukemia, and rheumatoid arthritis.

While CD20 itself is not an antigen, it can be recognized by the immune system as a foreign substance when a monoclonal antibody such as rituximab binds to it. This binding can trigger an immune response, leading to the destruction of the B-cells that express CD20 on their surface.

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.

Fetal blood refers to the blood circulating in a fetus during pregnancy. It is essential for the growth and development of the fetus, as it carries oxygen and nutrients from the placenta to the developing tissues and organs. Fetal blood also removes waste products, such as carbon dioxide, from the fetal tissues and transports them to the placenta for elimination.

Fetal blood has several unique characteristics that distinguish it from adult blood. For example, fetal hemoglobin (HbF) is the primary type of hemoglobin found in fetal blood, whereas adults primarily have adult hemoglobin (HbA). Fetal hemoglobin has a higher affinity for oxygen than adult hemoglobin, which allows it to more efficiently extract oxygen from the maternal blood in the placenta.

Additionally, fetal blood contains a higher proportion of reticulocytes (immature red blood cells) and nucleated red blood cells compared to adult blood. These differences reflect the high turnover rate of red blood cells in the developing fetus and the need for rapid growth and development.

Examination of fetal blood can provide important information about the health and well-being of the fetus during pregnancy. For example, fetal blood sampling (also known as cordocentesis or percutaneous umbilical blood sampling) can be used to diagnose genetic disorders, infections, and other conditions that may affect fetal development. However, this procedure carries risks, including preterm labor, infection, and fetal loss, and is typically only performed when there is a significant risk of fetal compromise or when other diagnostic tests have been inconclusive.

The Major Histocompatibility Complex (MHC) is a group of cell surface proteins in vertebrates that play a central role in the adaptive immune system. They are responsible for presenting peptide antigens to T-cells, which helps the immune system distinguish between self and non-self. The MHC is divided into two classes:

1. MHC Class I: These proteins present endogenous (intracellular) peptides to CD8+ T-cells (cytotoxic T-cells). The MHC class I molecule consists of a heavy chain and a light chain, together with an antigenic peptide.

2. MHC Class II: These proteins present exogenous (extracellular) peptides to CD4+ T-cells (helper T-cells). The MHC class II molecule is composed of two heavy chains and two light chains, together with an antigenic peptide.

MHC genes are highly polymorphic, meaning there are many different alleles within a population. This diversity allows for better recognition and presentation of various pathogens, leading to a more robust immune response. The term "histocompatibility" refers to the compatibility between donor and recipient MHC molecules in tissue transplantation. Incompatible MHC molecules can lead to rejection of the transplanted tissue due to an activated immune response against the foreign MHC antigens.

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.

An antigen is any substance that can stimulate an immune response, particularly the production of antibodies. Viral antigens are antigens that are found on or produced by viruses. They can be proteins, glycoproteins, or carbohydrates present on the surface or inside the viral particle.

Viral antigens play a crucial role in the immune system's recognition and response to viral infections. When a virus infects a host cell, it may display its antigens on the surface of the infected cell. This allows the immune system to recognize and target the infected cells for destruction, thereby limiting the spread of the virus.

Viral antigens are also important targets for vaccines. Vaccines typically work by introducing a harmless form of a viral antigen to the body, which then stimulates the production of antibodies and memory T-cells that can recognize and respond quickly and effectively to future infections with the actual virus.

It's worth noting that different types of viruses have different antigens, and these antigens can vary between strains of the same virus. This is why there are often different vaccines available for different viral diseases, and why flu vaccines need to be updated every year to account for changes in the circulating influenza virus strains.

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.

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.

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.

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.

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.

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.

A case-control study is an observational research design used to identify risk factors or causes of a disease or health outcome. In this type of study, individuals with the disease or condition (cases) are compared with similar individuals who do not have the disease or condition (controls). The exposure history or other characteristics of interest are then compared between the two groups to determine if there is an association between the exposure and the disease.

Case-control studies are often used when it is not feasible or ethical to conduct a randomized controlled trial, as they can provide valuable insights into potential causes of diseases or health outcomes in a relatively short period of time and at a lower cost than other study designs. However, because case-control studies rely on retrospective data collection, they are subject to biases such as recall bias and selection bias, which can affect the validity of the results. Therefore, it is important to carefully design and conduct case-control studies to minimize these potential sources of bias.

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.

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).

HLA (Human Leukocyte Antigen) antigens are a group of proteins found on the surface of cells in our body. They play a crucial role in the immune system's ability to differentiate between "self" and "non-self." HLA antigens are encoded by a group of genes located on chromosome 6, known as the major histocompatibility complex (MHC).

There are three types of HLA antigens: HLA class I, HLA class II, and HLA class III. HLA class I antigens are found on the surface of almost all cells in the body and help the immune system recognize and destroy virus-infected or cancerous cells. They consist of three components: HLA-A, HLA-B, and HLA-C.

HLA class II antigens are primarily found on the surface of immune cells, such as macrophages, B cells, and dendritic cells. They assist in the presentation of foreign particles (like bacteria and viruses) to CD4+ T cells, which then activate other parts of the immune system. HLA class II antigens include HLA-DP, HLA-DQ, and HLA-DR.

HLA class III antigens consist of various molecules involved in immune responses, such as cytokines and complement components. They are not directly related to antigen presentation.

The genetic diversity of HLA antigens is extensive, with thousands of variations or alleles. This diversity allows for a better ability to recognize and respond to a wide range of pathogens. However, this variation can also lead to compatibility issues in organ transplantation, as the recipient's immune system may recognize the donor's HLA antigens as foreign and attack the transplanted organ.

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.

"Macaca mulatta" is the scientific name for the Rhesus macaque, a species of monkey that is native to South, Central, and Southeast Asia. They are often used in biomedical research due to their genetic similarity to humans.

HLA-A antigens are a type of human leukocyte antigen (HLA) found on the surface of cells in our body. They are proteins that play an important role in the immune system by helping the body recognize and distinguish its own cells from foreign substances such as viruses, bacteria, and transplanted organs.

The HLA-A antigens are part of the major histocompatibility complex (MHC) class I molecules, which present peptide fragments from inside the cell to CD8+ T cells, also known as cytotoxic T lymphocytes (CTLs). The CTLs then recognize and destroy any cells that display foreign or abnormal peptides on their HLA-A antigens.

Each person has a unique set of HLA-A antigens, which are inherited from their parents. These antigens can vary widely between individuals, making it important to match HLA types in organ transplantation to reduce the risk of rejection. Additionally, certain HLA-A antigens have been associated with increased susceptibility or resistance to various diseases, including autoimmune disorders and infectious diseases.

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.

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.

"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.

Antibody specificity refers to the ability of an antibody to bind to a specific epitope or antigenic determinant on an antigen. Each antibody has a unique structure that allows it to recognize and bind to a specific region of an antigen, typically a small portion of the antigen's surface made up of amino acids or sugar residues. This highly specific binding is mediated by the variable regions of the antibody's heavy and light chains, which form a pocket that recognizes and binds to the epitope.

The specificity of an antibody is determined by its unique complementarity-determining regions (CDRs), which are loops of amino acids located in the variable domains of both the heavy and light chains. The CDRs form a binding site that recognizes and interacts with the epitope on the antigen. The precise fit between the antibody's binding site and the epitope is critical for specificity, as even small changes in the structure of either can prevent binding.

Antibody specificity is important in immune responses because it allows the immune system to distinguish between self and non-self antigens. This helps to prevent autoimmune reactions where the immune system attacks the body's own cells and tissues. Antibody specificity also plays a crucial role in diagnostic tests, such as ELISA assays, where antibodies are used to detect the presence of specific antigens in biological samples.

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.

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.

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.

Coxsackievirus infections are a type of viral illness caused by Coxsackie A and B viruses, which belong to the family Picornaviridae. These viruses can cause a wide range of symptoms, depending on the specific strain and the age and overall health of the infected individual.

The most common types of Coxsackievirus infections are hand, foot, and mouth disease (HFMD) and herpangina. HFMD is characterized by fever, sore throat, and a rash that typically appears on the hands, feet, and mouth. Herpangina is similar but is usually marked by painful sores in the back of the mouth or throat.

Other possible symptoms of Coxsackievirus infections include:

* Fever
* Headache
* Muscle aches
* Fatigue
* Nausea and vomiting
* Abdominal pain

In some cases, Coxsackievirus infections can lead to more serious complications, such as meningitis (inflammation of the membranes surrounding the brain and spinal cord), myocarditis (inflammation of the heart muscle), or pleurodynia (also known as "devil's grip," a painful inflammation of the chest and abdominal muscles).

Coxsackievirus infections are typically spread through close contact with an infected person, such as through respiratory droplets or by touching contaminated surfaces. The viruses can also be spread through fecal-oral transmission.

There is no specific treatment for Coxsackievirus infections, and most people recover on their own within a week or two. However, severe cases may require hospitalization and supportive care, such as fluids and pain relief. Prevention measures include good hygiene practices, such as washing hands frequently and avoiding close contact with sick individuals.

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.

Aging is a complex, progressive and inevitable process of bodily changes over time, characterized by the accumulation of cellular damage and degenerative changes that eventually lead to increased vulnerability to disease and death. It involves various biological, genetic, environmental, and lifestyle factors that contribute to the decline in physical and mental functions. The medical field studies aging through the discipline of gerontology, which aims to understand the underlying mechanisms of aging and develop interventions to promote healthy aging and extend the human healthspan.

Immunoglobulin A (IgA) is a type of antibody that plays a crucial role in the immune function of the human body. It is primarily found in external secretions, such as saliva, tears, breast milk, and sweat, as well as in mucous membranes lining the respiratory and gastrointestinal tracts. IgA exists in two forms: a monomeric form found in serum and a polymeric form found in secretions.

The primary function of IgA is to provide immune protection at mucosal surfaces, which are exposed to various environmental antigens, such as bacteria, viruses, parasites, and allergens. By doing so, it helps prevent the entry and colonization of pathogens into the body, reducing the risk of infections and inflammation.

IgA functions by binding to antigens present on the surface of pathogens or allergens, forming immune complexes that can neutralize their activity. These complexes are then transported across the epithelial cells lining mucosal surfaces and released into the lumen, where they prevent the adherence and invasion of pathogens.

In summary, Immunoglobulin A (IgA) is a vital antibody that provides immune defense at mucosal surfaces by neutralizing and preventing the entry of harmful antigens into the body.

ADP-ribosyl cyclase is an enzyme that catalyzes the conversion of nicotinamide adenine dinucleotide (NAD+) to cyclic ADP-ribose (cADPR). This enzyme plays a role in intracellular signaling, particularly in calcium mobilization in various cell types including immune cells and neurons. The regulation of this enzyme has been implicated in several physiological processes as well as in the pathophysiology of some diseases such as cancer and neurodegenerative disorders.

Autoantibodies are defined as antibodies that are produced by the immune system and target the body's own cells, tissues, or organs. These antibodies mistakenly identify certain proteins or molecules in the body as foreign invaders and attack them, leading to an autoimmune response. Autoantibodies can be found in various autoimmune diseases such as rheumatoid arthritis, lupus, and thyroiditis. The presence of autoantibodies can also be used as a diagnostic marker for certain conditions.

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.

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.

Antibodies, viral are proteins produced by the immune system in response to an infection with a virus. These antibodies are capable of recognizing and binding to specific antigens on the surface of the virus, which helps to neutralize or destroy the virus and prevent its replication. Once produced, these antibodies can provide immunity against future infections with the same virus.

Viral antibodies are typically composed of four polypeptide chains - two heavy chains and two light chains - that are held together by disulfide bonds. The binding site for the antigen is located at the tip of the Y-shaped structure, formed by the variable regions of the heavy and light chains.

There are five classes of antibodies in humans: IgA, IgD, IgE, IgG, and IgM. Each class has a different function and is distributed differently throughout the body. For example, IgG is the most common type of antibody found in the bloodstream and provides long-term immunity against viruses, while IgA is found primarily in mucous membranes and helps to protect against respiratory and gastrointestinal infections.

In addition to their role in the immune response, viral antibodies can also be used as diagnostic tools to detect the presence of a specific virus in a patient's blood or other bodily fluids.

CD38 is a type of antigen that is found on the surface of many different types of cells in the human body, including immune cells such as T-cells and B-cells. Antigens are substances (usually proteins) on the surface of cells that can be recognized by the immune system, triggering an immune response.

CD38 plays a role in several different cellular processes, including the regulation of calcium levels within cells, the production of energy in the form of ATP, and the modulation of immune responses. It is also involved in the activation and proliferation of T-cells and B-cells, which are critical components of the adaptive immune system.

CD38 can be targeted by certain types of immunotherapy, such as monoclonal antibodies, to help stimulate an immune response against cancer cells that express this antigen on their surface.

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.

Graft-versus-host disease (GVHD) is a condition that can occur after an allogeneic hematopoietic stem cell transplantation (HSCT), where the donated immune cells (graft) recognize the recipient's tissues (host) as foreign and attack them. This results in inflammation and damage to various organs, particularly the skin, gastrointestinal tract, and liver.

Acute GVHD typically occurs within 100 days of transplantation and is characterized by symptoms such as rash, diarrhea, and liver dysfunction. Chronic GVHD, on the other hand, can occur after 100 days or even years post-transplant and may present with a wider range of symptoms, including dry eyes and mouth, skin changes, lung involvement, and issues with mobility and flexibility in joints.

GVHD is a significant complication following allogeneic HSCT and can have a substantial impact on the patient's quality of life and overall prognosis. Preventative measures, such as immunosuppressive therapy, are often taken to reduce the risk of GVHD, but its management remains a challenge in transplant medicine.

Medical Definition of "Herpesvirus 4, Human" (Epstein-Barr Virus)

"Herpesvirus 4, Human," also known as Epstein-Barr virus (EBV), is a member of the Herpesviridae family and is one of the most common human viruses. It is primarily transmitted through saliva and is often referred to as the "kissing disease."

EBV is the causative agent of infectious mononucleosis (IM), also known as glandular fever, which is characterized by symptoms such as fatigue, sore throat, fever, and swollen lymph nodes. The virus can also cause other diseases, including certain types of cancer, such as Burkitt's lymphoma, Hodgkin's lymphoma, and nasopharyngeal carcinoma.

Once a person becomes infected with EBV, the virus remains in the body for the rest of their life, residing in certain white blood cells called B lymphocytes. In most people, the virus remains dormant and does not cause any further symptoms. However, in some individuals, the virus may reactivate, leading to recurrent or persistent symptoms.

EBV infection is diagnosed through various tests, including blood tests that detect antibodies against the virus or direct detection of the virus itself through polymerase chain reaction (PCR) assays. There is no cure for EBV infection, and treatment is generally supportive, focusing on relieving symptoms and managing complications. Prevention measures include practicing good hygiene, avoiding close contact with infected individuals, and not sharing personal items such as toothbrushes or drinking glasses.

Viremia is a medical term that refers to the presence of viruses in the bloodstream. It occurs when a virus successfully infects a host and replicates within the body's cells, releasing new viral particles into the blood. This condition can lead to various clinical manifestations depending on the specific virus involved and the immune response of the infected individual. Some viral infections result in asymptomatic viremia, while others can cause severe illness or even life-threatening conditions. The detection of viremia is crucial for diagnosing certain viral infections and monitoring disease progression or treatment effectiveness.

Viral load refers to the amount or quantity of virus (like HIV, Hepatitis C, SARS-CoV-2) present in an individual's blood or bodily fluids. It is often expressed as the number of virus copies per milliliter of blood or fluid. Monitoring viral load is important in managing and treating certain viral infections, as a higher viral load may indicate increased infectivity, disease progression, or response to treatment.

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.

Systemic Lupus Erythematosus (SLE) is a complex autoimmune disease that can affect almost any organ or system in the body. In SLE, the immune system produces an exaggerated response, leading to the production of autoantibodies that attack the body's own cells and tissues, causing inflammation and damage. The symptoms and severity of SLE can vary widely from person to person, but common features include fatigue, joint pain, skin rashes (particularly a "butterfly" rash across the nose and cheeks), fever, hair loss, and sensitivity to sunlight.

Systemic lupus erythematosus can also affect the kidneys, heart, lungs, brain, blood vessels, and other organs, leading to a wide range of symptoms such as kidney dysfunction, chest pain, shortness of breath, seizures, and anemia. The exact cause of SLE is not fully understood, but it is believed to involve a combination of genetic, environmental, and hormonal factors. Treatment typically involves medications to suppress the immune system and manage symptoms, and may require long-term management by a team of healthcare professionals.

Graft rejection is an immune response that occurs when transplanted tissue or organ (the graft) is recognized as foreign by the recipient's immune system, leading to the activation of immune cells to attack and destroy the graft. This results in the failure of the transplant and the need for additional medical intervention or another transplant. There are three types of graft rejection: hyperacute, acute, and chronic. Hyperacute rejection occurs immediately or soon after transplantation due to pre-existing antibodies against the graft. Acute rejection typically occurs within weeks to months post-transplant and is characterized by the infiltration of T-cells into the graft. Chronic rejection, which can occur months to years after transplantation, is a slow and progressive process characterized by fibrosis and tissue damage due to ongoing immune responses against the graft.

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.

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.

HIV seronegativity is a term used to describe a person who has tested negative for HIV (Human Immunodeficiency Virus) antibodies in their blood. This means that the individual does not show evidence of current or past infection with HIV, which can cause AIDS (Acquired Immune Deficiency Syndrome). However, it's important to note that there is a window period after initial infection during which a person may test negative for HIV antibodies, even though they are indeed infected. This window period typically lasts between 2-6 weeks but can extend up to 3 months in some cases. Therefore, if someone believes they have been exposed to HIV, they should consider getting tested again after this window period has passed.

Autoimmune thyroiditis, also known as Hashimoto's disease, is a chronic inflammation of the thyroid gland caused by an autoimmune response. In this condition, the immune system produces antibodies that attack and damage the thyroid gland, leading to hypothyroidism (underactive thyroid). The thyroid gland may become enlarged (goiter), and symptoms can include fatigue, weight gain, cold intolerance, constipation, dry skin, and depression. Autoimmune thyroiditis is more common in women than men and tends to run in families. It is often associated with other autoimmune disorders such as rheumatoid arthritis, Addison's disease, and type 1 diabetes. The diagnosis is typically made through blood tests that measure levels of thyroid hormones and antibodies. Treatment usually involves thyroid hormone replacement therapy to manage the symptoms of hypothyroidism.

'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.

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.

Forkhead transcription factors (FOX) are a family of proteins that play crucial roles in the regulation of gene expression through the process of binding to specific DNA sequences, thereby controlling various biological processes such as cell growth, differentiation, and apoptosis. These proteins are characterized by a conserved DNA-binding domain, known as the forkhead box or FOX domain, which adopts a winged helix structure that recognizes and binds to the consensus sequence 5'-(G/A)(T/C)AA(C/A)A-3'.

The FOX family is further divided into subfamilies based on the structure of their DNA-binding domains, with each subfamily having distinct functions. For example, FOXP proteins are involved in brain development and function, while FOXO proteins play a key role in regulating cellular responses to stress and metabolism. Dysregulation of forkhead transcription factors has been implicated in various diseases, including cancer, diabetes, and neurodegenerative disorders.

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.

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.

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.

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.

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.

The thoracic duct is the largest lymphatic vessel in the human body. It is a part of the lymphatic system, which helps to regulate fluid balance and immune function. The thoracic duct originates from the cisterna chyli, a dilated sac located in the abdomen near the aorta.

The thoracic duct collects lymph from the lower extremities, abdomen, pelvis, and left side of the thorax (chest). It ascends through the diaphragm and enters the chest, where it passes through the mediastinum (the central part of the chest between the lungs) and eventually drains into the left subclavian vein.

The thoracic duct plays a crucial role in transporting lymphatic fluid, which contains white blood cells, fats, proteins, and other substances, back into the circulatory system. Any obstruction or damage to the thoracic duct can lead to lymph accumulation in the surrounding tissues, causing swelling and other symptoms.

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.

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.

Humoral immunity is a type of immune response in which the body produces proteins called antibodies that circulate in bodily fluids such as blood and help to protect against infection. This form of immunity involves the interaction between antigens (foreign substances that trigger an immune response) and soluble factors, including antibodies, complement proteins, and cytokines.

When a pathogen enters the body, it is recognized as foreign by the immune system, which triggers the production of specific antibodies to bind to and neutralize or destroy the pathogen. These antibodies are produced by B cells, a type of white blood cell that is part of the adaptive immune system.

Humoral immunity provides protection against extracellular pathogens, such as bacteria and viruses, that exist outside of host cells. It is an important component of the body's defense mechanisms and plays a critical role in preventing and fighting off 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.

Cytomegalovirus (CMV) infections are caused by the human herpesvirus 5 (HHV-5), a type of herpesvirus. The infection can affect people of all ages, but it is more common in individuals with weakened immune systems, such as those with HIV/AIDS or who have undergone organ transplantation.

CMV can be spread through close contact with an infected person's saliva, urine, blood, tears, semen, or breast milk. It can also be spread through sexual contact or by sharing contaminated objects, such as toys, eating utensils, or drinking glasses. Once a person is infected with CMV, the virus remains in their body for life and can reactivate later, causing symptoms to recur.

Most people who are infected with CMV do not experience any symptoms, but some may develop a mononucleosis-like illness, characterized by fever, fatigue, swollen glands, and sore throat. In people with weakened immune systems, CMV infections can cause more severe symptoms, including pneumonia, gastrointestinal disease, retinitis, and encephalitis.

Congenital CMV infection occurs when a pregnant woman passes the virus to her fetus through the placenta. This can lead to serious complications, such as hearing loss, vision loss, developmental delays, and mental disability.

Diagnosis of CMV infections is typically made through blood tests or by detecting the virus in bodily fluids, such as urine or saliva. Treatment depends on the severity of the infection and the patient's overall health. Antiviral medications may be prescribed to help manage symptoms and prevent complications.

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.

CTLA-4 (Cytotoxic T-Lymphocyte Associated Protein 4) antigen is a type of protein found on the surface of activated T cells, which are a type of white blood cell in the immune system. CTLA-4 plays an important role in regulating the immune response by functioning as a negative regulator of T cell activation.

CTLA-4 binds to CD80 and CD86 molecules on the surface of antigen-presenting cells, which are cells that display foreign antigens to T cells and activate them. By binding to these molecules, CTLA-4 inhibits T cell activation and helps prevent an overactive immune response.

CTLA-4 is a target for cancer immunotherapy because blocking its function can enhance the anti-tumor immune response. Certain drugs called checkpoint inhibitors work by blocking CTLA-4, allowing T cells to remain active and attack tumor cells more effectively.

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.

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.

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.

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.

Myocarditis is an inflammation of the myocardium, which is the middle layer of the heart wall. The myocardium is composed of cardiac muscle cells and is responsible for the heart's pumping function. Myocarditis can be caused by various infectious and non-infectious agents, including viruses, bacteria, fungi, parasites, autoimmune diseases, toxins, and drugs.

In myocarditis, the inflammation can damage the cardiac muscle cells, leading to decreased heart function, arrhythmias (irregular heart rhythms), and in severe cases, heart failure or even sudden death. Symptoms of myocarditis may include chest pain, shortness of breath, fatigue, palpitations, and swelling in the legs, ankles, or abdomen.

The diagnosis of myocarditis is often based on a combination of clinical presentation, laboratory tests, electrocardiogram (ECG), echocardiography, cardiac magnetic resonance imaging (MRI), and endomyocardial biopsy. Treatment depends on the underlying cause and severity of the disease and may include medications to support heart function, reduce inflammation, control arrhythmias, and prevent further damage to the heart muscle. In some cases, hospitalization and intensive care may be necessary.

Lymphoma is a type of cancer that originates from the white blood cells called lymphocytes, which are part of the immune system. These cells are found in various parts of the body such as the lymph nodes, spleen, bone marrow, and other organs. Lymphoma can be classified into two main types: Hodgkin lymphoma (HL) and non-Hodgkin lymphoma (NHL).

HL is characterized by the presence of a specific type of abnormal lymphocyte called Reed-Sternberg cells, while NHL includes a diverse group of lymphomas that lack these cells. The symptoms of lymphoma may include swollen lymph nodes, fever, night sweats, weight loss, and fatigue.

The exact cause of lymphoma is not known, but it is believed to result from genetic mutations in the lymphocytes that lead to uncontrolled cell growth and division. Exposure to certain viruses, chemicals, and radiation may increase the risk of developing lymphoma. Treatment options for lymphoma depend on various factors such as the type and stage of the disease, age, and overall health of the patient. Common treatments include chemotherapy, radiation therapy, immunotherapy, and stem cell transplantation.

A provirus is a form of the genetic material of a retrovirus that is integrated into the DNA of the host cell it has infected. Once integrated, the provirus is replicated along with the host's own DNA every time the cell divides, and it becomes a permanent part of the host's genome.

The process of integration involves the reverse transcription of the retroviral RNA genome into DNA by the enzyme reverse transcriptase, followed by the integration of the resulting double-stranded proviral DNA into the host chromosome by the enzyme integrase.

Proviruses can remain dormant and inactive for long periods of time, or they can become active and produce new viral particles that can infect other cells. In some cases, proviruses can also disrupt the normal functioning of host genes, leading to various diseases such as cancer.

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.

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.

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.

Passive immunization is a type of temporary immunity that is transferred to an individual through the injection of antibodies produced outside of the body, rather than through the active production of antibodies in the body in response to vaccination or infection. This can be done through the administration of preformed antibodies, such as immune globulins, which contain a mixture of antibodies that provide immediate protection against specific diseases.

Passive immunization is often used in situations where individuals have been exposed to a disease and do not have time to develop their own active immune response, or in cases where individuals are unable to produce an adequate immune response due to certain medical conditions. It can also be used as a short-term measure to provide protection until an individual can receive a vaccination that will confer long-term immunity.

Passive immunization provides immediate protection against disease, but the protection is typically short-lived, lasting only a few weeks or months. This is because the transferred antibodies are gradually broken down and eliminated by the body over time. In contrast, active immunization confers long-term immunity through the production of memory cells that can mount a rapid and effective immune response upon re-exposure to the same pathogen in the future.

The Yellow Fever Vaccine is a vaccine that protects against the yellow fever virus, which is transmitted to humans through the bites of infected mosquitoes. The vaccine contains live, weakened yellow fever virus, and it works by stimulating the immune system to produce an immune response that provides protection against the disease.

The yellow fever vaccine is recommended for people who are traveling to areas where yellow fever is common, including parts of Africa and South America. It is also required for entry into some countries in these regions. The vaccine is generally safe and effective, but it can cause mild side effects such as headache, muscle pain, and fever in some people. Serious side effects are rare, but may include allergic reactions or infection with the weakened virus used in the vaccine.

It's important to note that yellow fever vaccine may not be recommended for certain individuals, including infants younger than 6 months, pregnant women, people with weakened immune systems, and those with a history of severe allergic reaction to a previous dose of the vaccine or any component of the vaccine. It is always best to consult with a healthcare provider before receiving any vaccination.

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.

Hydrocortisone is a synthetic glucocorticoid, which is a class of steroid hormones. It is identical to the naturally occurring cortisol, a hormone produced by the adrenal gland that helps regulate metabolism and helps your body respond to stress. Hydrocortisone has anti-inflammatory effects and is used to treat various inflammatory conditions such as allergies, skin disorders, and autoimmune diseases. It works by suppressing the immune system's response to reduce swelling, redness, itching, and other symptoms caused by inflammation.

Hydrocortisone is available in different forms, including oral tablets, topical creams, lotions, gels, and ointments, as well as injectable solutions. The specific use and dosage depend on the condition being treated and the individual patient's medical history and current health status.

As with any medication, hydrocortisone can have side effects, especially when used in high doses or for extended periods. Common side effects include increased appetite, weight gain, mood changes, insomnia, and skin thinning. Long-term use of hydrocortisone may also increase the risk of developing osteoporosis, diabetes, cataracts, and other health problems. Therefore, it is essential to follow your healthcare provider's instructions carefully when using this medication.

Enterovirus B, Human (HEVB) is a type of enterovirus that infects humans. Enteroviruses are small viruses that belong to the Picornaviridae family and are named after the Greek word "pico" meaning small. They are further classified into several species, including Human Enterovirus B (HEV-B).

HEVB includes several serotypes, such as Coxsackievirus A9, A16, and B types, and Echoviruses. These viruses are typically transmitted through the fecal-oral route or respiratory droplets and can cause a range of illnesses, from mild symptoms like fever, rash, and sore throat to more severe diseases such as meningitis, myocarditis, and paralysis.

HEVB infections are common worldwide, and people of all ages can be affected. However, young children and individuals with weakened immune systems are at higher risk for severe illness. Prevention measures include good hygiene practices, such as washing hands frequently and avoiding close contact with sick individuals. There is no specific treatment for HEVB infections, and most cases resolve on their own within a few days to a week. However, hospitalization may be necessary for severe cases.

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.

The Receptor-CD3 Complex is a multimeric protein complex found on the surface of T-cells, a type of white blood cell crucial to the adaptive immune system. The complex plays a critical role in the activation and regulation of T-cells. It is composed of the T-cell receptor (TCR) and the CD3 proteins (CD3δ, ε, γ, and ζ).

The T-cell receptor is responsible for recognizing specific antigens presented in the context of major histocompatibility complex (MHC) molecules on the surface of antigen-presenting cells. The CD3 proteins are involved in signal transduction upon TCR engagement with an antigen, leading to T-cell activation and downstream effects such as cytokine production and cytotoxicity.

An antigen is any substance (usually a protein) that can be recognized by the immune system and stimulate an immune response. Antigens are typically foreign substances, but they can also include self-proteins in certain circumstances, such as during autoimmune diseases. In the context of T-cells, antigens are presented in the form of peptides bound to MHC molecules on the surface of antigen-presenting cells.

T-cells are a type of lymphocyte that plays a central role in cell-mediated immunity. They recognize and respond to specific antigens, contributing to the elimination of infected or damaged cells and providing long-lasting immune protection against pathogens. T-cells can be further classified into various subsets based on their surface receptors and functions, including CD4+ helper T-cells, CD8+ cytotoxic T-cells, regulatory T-cells, and memory T-cells.

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.

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.

A "Graft versus Host Reaction" (GVHR) is a condition that can occur after an organ or bone marrow transplant, where the immune cells in the graft (transplanted tissue) recognize and attack the recipient's (host's) tissues as foreign. This reaction occurs because the donor's immune cells (graft) are able to recognize the host's cells as different from their own due to differences in proteins called human leukocyte antigens (HLAs).

The GVHR can affect various organs, including the skin, liver, gastrointestinal tract, and lungs. Symptoms may include rash, diarrhea, jaundice, and respiratory distress. The severity of the reaction can vary widely, from mild to life-threatening.

To prevent or reduce the risk of GVHR, immunosuppressive drugs are often given to the recipient before and after transplantation to suppress their immune system and prevent it from attacking the graft. Despite these measures, GVHR can still occur in some cases, particularly when there is a significant mismatch between the donor and recipient HLAs.

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.

Muromonab-CD3 is a type of immunosuppressant medication that is used in the treatment of acute organ rejection in patients who have received organ transplants. It is a monoclonal antibody that specifically targets and binds to the CD3 receptor found on the surface of T-lymphocytes, which are a type of white blood cell that plays a central role in the immune response.

By binding to the CD3 receptor, Muromonab-CD3 inhibits the activation and proliferation of T-lymphocytes, thereby suppressing the immune system's ability to recognize and attack the transplanted organ. This helps to prevent or reverse the process of acute organ rejection.

Muromonab-CD3 is administered intravenously and is typically given as a series of doses over several days. It may be used in combination with other immunosuppressive drugs to achieve optimal results. As with any medication, Muromonab-CD3 can have side effects, including fever, chills, nausea, and headache. More serious side effects, such as anaphylaxis or severe infections, may also occur, and patients should be closely monitored during treatment.

A chronic disease is a long-term medical condition that often progresses slowly over a period of years and requires ongoing management and care. These diseases are typically not fully curable, but symptoms can be managed to improve quality of life. Common chronic diseases include heart disease, stroke, cancer, diabetes, arthritis, and COPD (chronic obstructive pulmonary disease). They are often associated with advanced age, although they can also affect children and younger adults. Chronic diseases can have significant impacts on individuals' physical, emotional, and social well-being, as well as on healthcare systems and society at large.

Thymectomy is a surgical procedure that involves the removal of the thymus gland. The thymus gland is a part of the immune system located in the upper chest, behind the sternum (breastbone), and above the heart. It is responsible for producing white blood cells called T-lymphocytes, which help fight infections.

Thymectomy is often performed as a treatment option for patients with certain medical conditions, such as:

* Myasthenia gravis: an autoimmune disorder that causes muscle weakness and fatigue. In some cases, the thymus gland may contain abnormal cells that contribute to the development of myasthenia gravis. Removing the thymus gland can help improve symptoms in some patients with this condition.
* Thymomas: tumors that develop in the thymus gland. While most thymomas are benign (non-cancerous), some can be malignant (cancerous) and may require surgical removal.
* Myasthenic syndrome: a group of disorders characterized by muscle weakness and fatigue, similar to myasthenia gravis. In some cases, the thymus gland may be abnormal and contribute to the development of these conditions. Removing the thymus gland can help improve symptoms in some patients.

Thymectomy can be performed using various surgical approaches, including open surgery (through a large incision in the chest), video-assisted thoracoscopic surgery (VATS, using small incisions and a camera to guide the procedure), or robotic-assisted surgery (using a robot to perform the procedure through small incisions). The choice of surgical approach depends on several factors, including the size and location of the thymus gland, the patient's overall health, and the surgeon's expertise.

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.

Infection is defined medically as the invasion and multiplication of pathogenic microorganisms such as bacteria, viruses, fungi, or parasites within the body, which can lead to tissue damage, illness, and disease. This process often triggers an immune response from the host's body in an attempt to eliminate the infectious agents and restore homeostasis. Infections can be transmitted through various routes, including airborne particles, direct contact with contaminated surfaces or bodily fluids, sexual contact, or vector-borne transmission. The severity of an infection may range from mild and self-limiting to severe and life-threatening, depending on factors such as the type and quantity of pathogen, the host's immune status, and any underlying health conditions.

Prednisolone is a synthetic glucocorticoid drug, which is a class of steroid hormones. It is commonly used in the treatment of various inflammatory and autoimmune conditions due to its potent anti-inflammatory and immunosuppressive effects. Prednisolone works by binding to specific receptors in cells, leading to changes in gene expression that reduce the production of substances involved in inflammation, such as cytokines and prostaglandins.

Prednisolone is available in various forms, including tablets, syrups, and injectable solutions. It can be used to treat a wide range of medical conditions, including asthma, rheumatoid arthritis, inflammatory bowel disease, allergies, skin conditions, and certain types of cancer.

Like other steroid medications, prednisolone can have significant side effects if used in high doses or for long periods of time. These may include weight gain, mood changes, increased risk of infections, osteoporosis, diabetes, and adrenal suppression. As a result, the use of prednisolone should be closely monitored by a healthcare professional to ensure that its benefits outweigh its risks.

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).

Immunoconjugates are biomolecules created by the conjugation (coupling) of an antibody or antibody fragment with a cytotoxic agent, such as a drug, radionuclide, or toxin. This coupling is designed to direct the cytotoxic agent specifically to target cells, usually cancer cells, against which the antibody is directed, thereby increasing the effectiveness and reducing the side effects of the therapy.

The antibody part of the immunoconjugate recognizes and binds to specific antigens (proteins or other molecules) on the surface of the target cells, while the cytotoxic agent part enters the cell and disrupts its function, leading to cell death. The linker between the two parts is designed to be stable in circulation but can release the cytotoxic agent once inside the target cell.

Immunoconjugates are a promising area of research in targeted cancer therapy, as they offer the potential for more precise and less toxic treatments compared to traditional chemotherapy. However, their development and use also pose challenges, such as ensuring that the immunoconjugate binds specifically to the target cells and not to normal cells, optimizing the dose and schedule of treatment, and minimizing the risk of resistance to the therapy.

Immunologic capping is a biological process that occurs in immune cells, particularly B lymphocytes and neutrophils. It refers to the redistribution and clustering of immunoglobulin receptors or antibodies on the cell surface upon engagement with their specific antigens. This phenomenon leads to the formation of a cap-like structure at one pole of the cell, which is then internalized by endocytosis, followed by the degradation of the antigen-antibody complex in lysosomes. Immunologic capping helps regulate immune responses and contributes to the elimination of antigens from the cell surface.

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.

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.

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.

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.

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.

A genetic vector is a vehicle, often a plasmid or a virus, that is used to introduce foreign DNA into a host cell as part of genetic engineering or gene therapy techniques. The vector contains the desired gene or genes, along with regulatory elements such as promoters and enhancers, which are needed for the expression of the gene in the target cells.

The choice of vector depends on several factors, including the size of the DNA to be inserted, the type of cell to be targeted, and the efficiency of uptake and expression required. Commonly used vectors include plasmids, adenoviruses, retroviruses, and lentiviruses.

Plasmids are small circular DNA molecules that can replicate independently in bacteria. They are often used as cloning vectors to amplify and manipulate DNA fragments. Adenoviruses are double-stranded DNA viruses that infect a wide range of host cells, including human cells. They are commonly used as gene therapy vectors because they can efficiently transfer genes into both dividing and non-dividing cells.

Retroviruses and lentiviruses are RNA viruses that integrate their genetic material into the host cell's genome. This allows for stable expression of the transgene over time. Lentiviruses, a subclass of retroviruses, have the advantage of being able to infect non-dividing cells, making them useful for gene therapy applications in post-mitotic tissues such as neurons and muscle cells.

Overall, genetic vectors play a crucial role in modern molecular biology and medicine, enabling researchers to study gene function, develop new therapies, and modify organisms for various purposes.

An antigen-antibody reaction is a specific immune response that occurs when an antigen (a foreign substance, such as a protein or polysaccharide on the surface of a bacterium or virus) comes into contact with a corresponding antibody (a protective protein produced by the immune system in response to the antigen). The antigen and antibody bind together, forming an antigen-antibody complex. This interaction can neutralize the harmful effects of the antigen, mark it for destruction by other immune cells, or activate complement proteins to help eliminate the antigen from the body. Antigen-antibody reactions are a crucial part of the adaptive immune response and play a key role in the body's defense against infection and disease.

The intestines, also known as the bowel, are a part of the digestive system that extends from the stomach to the anus. They are responsible for the further breakdown and absorption of nutrients from food, as well as the elimination of waste products. The intestines can be divided into two main sections: the small intestine and the large intestine.

The small intestine is a long, coiled tube that measures about 20 feet in length and is lined with tiny finger-like projections called villi, which increase its surface area and enhance nutrient absorption. The small intestine is where most of the digestion and absorption of nutrients takes place.

The large intestine, also known as the colon, is a wider tube that measures about 5 feet in length and is responsible for absorbing water and electrolytes from digested food, forming stool, and eliminating waste products from the body. The large intestine includes several regions, including the cecum, colon, rectum, and anus.

Together, the intestines play a critical role in maintaining overall health and well-being by ensuring that the body receives the nutrients it needs to function properly.

Hematopoietic Stem Cell Transplantation (HSCT) is a medical procedure where hematopoietic stem cells (immature cells that give rise to all blood cell types) are transplanted into a patient. This procedure is often used to treat various malignant and non-malignant disorders affecting the hematopoietic system, such as leukemias, lymphomas, multiple myeloma, aplastic anemia, inherited immune deficiency diseases, and certain genetic metabolic disorders.

The transplantation can be autologous (using the patient's own stem cells), allogeneic (using stem cells from a genetically matched donor, usually a sibling or unrelated volunteer), or syngeneic (using stem cells from an identical twin).

The process involves collecting hematopoietic stem cells, most commonly from the peripheral blood or bone marrow. The collected cells are then infused into the patient after the recipient's own hematopoietic system has been ablated (or destroyed) using high-dose chemotherapy and/or radiation therapy. This allows the donor's stem cells to engraft, reconstitute, and restore the patient's hematopoietic system.

HSCT is a complex and potentially risky procedure with various complications, including graft-versus-host disease, infections, and organ damage. However, it offers the potential for cure or long-term remission in many patients with otherwise fatal diseases.

Simian Immunodeficiency Virus (SIV) is a retrovirus that primarily infects African non-human primates and is the direct ancestor of Human Immunodeficiency Virus type 2 (HIV-2). It is similar to HIV in its structure, replication strategy, and ability to cause an immunodeficiency disease in its host. SIV infection in its natural hosts is typically asymptomatic and non-lethal, but it can cause AIDS-like symptoms in other primate species. Research on SIV in its natural hosts has provided valuable insights into the mechanisms of HIV pathogenesis and potential strategies for prevention and treatment of AIDS.

Yellow fever is an acute viral hemorrhagic disease that's transmitted to humans through the bite of infected mosquitoes. The "yellow" in the name refers to the jaundice that can occur in some patients, resulting from liver damage caused by the virus. The disease is endemic in tropical regions of Africa and Central and South America.

The yellow fever virus is a single-stranded RNA virus that belongs to the Flaviviridae family, genus Flavivirus. It's closely related to other mosquito-borne viruses like dengue and Zika. The virus has three distinct geographical variants (West African, East African, and South American), each with different epidemiological patterns and clinical features.

The incubation period for yellow fever is typically 3 to 6 days after infection. The initial symptoms include fever, chills, headache, back pain, myalgia, and fatigue. Most patients recover after this initial phase, but around 15% of those infected enter a more severe phase characterized by high fever, jaundice, abdominal pain, vomiting, bleeding, and often rapid death within 7 to 10 days.

There is no specific treatment for yellow fever, and management is focused on supportive care, including fluid replacement, blood transfusions, and addressing any complications that arise. Prevention relies on vaccination and mosquito control measures. The yellow fever vaccine is safe and highly effective, providing immunity in 95% of those who receive it. A single dose offers lifelong protection in most individuals. Mosquito control efforts, such as reducing breeding sites and using insecticide-treated materials, can help prevent the spread of the virus in affected areas.

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.

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.

Chromium radioisotopes are unstable isotopes or variants of the chemical element chromium that emit radiation as they decay into more stable forms. These isotopes have an excess of energy and particles, making them unstable and capable of emitting ionizing radiation in the form of gamma rays or subatomic particles such as alpha or beta particles.

Chromium has several radioisotopes, including chromium-50, chromium-51, and chromium-53, among others. Chromium-51 is one of the most commonly used radioisotopes in medical applications, particularly in diagnostic procedures such as red blood cell labeling and imaging studies.

It's important to note that handling and using radioisotopes require proper training and safety measures due to their potential radiation hazards.

HIV seropositivity is a term used to describe a positive result on an HIV antibody test. This means that the individual has developed antibodies against the Human Immunodeficiency Virus (HIV), indicating that they have been infected with the virus. However, it's important to note that this does not necessarily mean that the person has AIDS, as there can be a long period between HIV infection and the development of AIDS.

Ionomycin is not a medical term per se, but it is a chemical compound used in medical and biological research. Ionomycin is a type of ionophore, which is a molecule that can transport ions across cell membranes. Specifically, ionomycin is known to transport calcium ions (Ca²+).

In medical research, ionomycin is often used to study the role of calcium in various cellular processes, such as signal transduction, gene expression, and muscle contraction. It can be used to selectively increase intracellular calcium concentrations in experiments, allowing researchers to observe the effects on cell function. Ionomycin is also used in the study of calcium-dependent enzymes and channels.

It's important to note that ionomycin is not used as a therapeutic agent in clinical medicine due to its potential toxicity and narrow range of applications.

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.

Isoantigens are antigens that are present on the cells or tissues of one individual of a species, but are absent or different in another individual of the same species. They are also known as "alloantigens." Isoantigens are most commonly found on the surface of red blood cells and other tissues, and they can stimulate an immune response when transplanted into a different individual. This is because the recipient's immune system recognizes the isoantigens as foreign and mounts a defense against them. Isoantigens are important in the field of transplantation medicine, as they must be carefully matched between donor and recipient to reduce the risk of rejection.

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

Cross reactions, in the context of medical diagnostics and immunology, refer to a situation where an antibody or a immune response directed against one antigen also reacts with a different antigen due to similarities in their molecular structure. This can occur in allergy testing, where a person who is allergic to a particular substance may have a positive test result for a different but related substance because of cross-reactivity between them. For example, some individuals who are allergic to birch pollen may also have symptoms when eating certain fruits, such as apples, due to cross-reactive proteins present in both.

A newborn infant is a baby who is within the first 28 days of life. This period is also referred to as the neonatal period. Newborns require specialized care and attention due to their immature bodily systems and increased vulnerability to various health issues. They are closely monitored for signs of well-being, growth, and development during this critical time.

Antibody-producing cells, also known as plasma cells, are a type of white blood cell that is responsible for producing and secreting antibodies in response to a foreign substance or antigen. These cells are derived from B lymphocytes, which become activated upon encountering an antigen and differentiate into plasma cells.

Once activated, plasma cells can produce large amounts of specific antibodies that bind to the antigen, marking it for destruction by other immune cells. Antibody-producing cells play a crucial role in the body's humoral immune response, which helps protect against infection and disease.

Nonparametric statistics is a branch of statistics that does not rely on assumptions about the distribution of variables in the population from which the sample is drawn. In contrast to parametric methods, nonparametric techniques make fewer assumptions about the data and are therefore more flexible in their application. Nonparametric tests are often used when the data do not meet the assumptions required for parametric tests, such as normality or equal variances.

Nonparametric statistical methods include tests such as the Wilcoxon rank-sum test (also known as the Mann-Whitney U test) for comparing two independent groups, the Wilcoxon signed-rank test for comparing two related groups, and the Kruskal-Wallis test for comparing more than two independent groups. These tests use the ranks of the data rather than the actual values to make comparisons, which allows them to be used with ordinal or continuous data that do not meet the assumptions of parametric tests.

Overall, nonparametric statistics provide a useful set of tools for analyzing data in situations where the assumptions of parametric methods are not met, and can help researchers draw valid conclusions from their data even when the data are not normally distributed or have other characteristics that violate the assumptions of parametric tests.

Cancer vaccines are a type of immunotherapy that stimulate the body's own immune system to recognize and destroy cancer cells. They can be prophylactic (preventive) or therapeutic (treatment) in nature. Prophylactic cancer vaccines, such as the human papillomavirus (HPV) vaccine, are designed to prevent the initial infection that can lead to certain types of cancer. Therapeutic cancer vaccines, on the other hand, are used to treat existing cancer by boosting the immune system's ability to identify and eliminate cancer cells. These vaccines typically contain specific antigens (proteins or sugars) found on the surface of cancer cells, which help the immune system to recognize and target them.

It is important to note that cancer vaccines are different from vaccines used to prevent infectious diseases, such as measles or influenza. While traditional vaccines introduce a weakened or inactivated form of a virus or bacteria to stimulate an immune response, cancer vaccines focus on training the immune system to recognize and attack cancer cells specifically.

There are several types of cancer vaccines under investigation, including:

1. Autologous cancer vaccines: These vaccines use the patient's own tumor cells, which are processed and then reintroduced into the body to stimulate an immune response.
2. Peptide-based cancer vaccines: These vaccines contain specific pieces (peptides) of proteins found on the surface of cancer cells. They are designed to trigger an immune response against cells that express these proteins.
3. Dendritic cell-based cancer vaccines: Dendritic cells are a type of immune cell responsible for presenting antigens to other immune cells, activating them to recognize and destroy infected or cancerous cells. In this approach, dendritic cells are isolated from the patient's blood, exposed to cancer antigens in the lab, and then reintroduced into the body to stimulate an immune response.
4. DNA-based cancer vaccines: These vaccines use pieces of DNA that code for specific cancer antigens. Once inside the body, these DNA fragments are taken up by cells, leading to the production of the corresponding antigen and triggering an immune response.
5. Viral vector-based cancer vaccines: In this approach, a harmless virus is modified to carry genetic material encoding cancer antigens. When introduced into the body, the virus infects cells, causing them to produce the cancer antigen and stimulating an immune response.

While some cancer vaccines have shown promising results in clinical trials, none have yet been approved for widespread use by regulatory authorities such as the US Food and Drug Administration (FDA). Researchers continue to explore and refine various vaccine strategies to improve their efficacy and safety.

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.

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.

Interleukin-7 (IL-7) is a small signaling protein that is involved in the development and function of immune cells, particularly T cells and B cells. It is produced by stromal cells found in the bone marrow, thymus, and lymphoid organs. IL-7 binds to its receptor, IL-7R, which is expressed on the surface of immature T cells and B cells, as well as some mature immune cells.

IL-7 plays a critical role in the survival, proliferation, and differentiation of T cells and B cells during their development in the thymus and bone marrow, respectively. It also helps to maintain the homeostasis of these cell populations in peripheral tissues by promoting their survival and preventing apoptosis.

In addition to its role in immune cell development and homeostasis, IL-7 has been shown to have potential therapeutic applications in the treatment of various diseases, including cancer, infectious diseases, and autoimmune disorders. However, further research is needed to fully understand its mechanisms of action and potential side effects before it can be widely used in clinical settings.

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.

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.

Interleukin-15 (IL-15) is a small protein with a molecular weight of approximately 14 to 15 kilodaltons. It belongs to the class of cytokines known as the four-alpha-helix bundle family, which also includes IL-2, IL-4, and IL-7.

IL-15 is primarily produced by monocytes, macrophages, and dendritic cells, but it can also be produced by other cell types such as fibroblasts, epithelial cells, and endothelial cells. It plays a crucial role in the immune system by regulating the activation, proliferation, and survival of various immune cells, including T cells, natural killer (NK) cells, and dendritic cells.

IL-15 binds to its receptor complex, which consists of three components: IL-15Rα, IL-2/IL-15Rβ, and the common γ-chain (γc). The binding of IL-15 to this receptor complex leads to the activation of several signaling pathways, including the JAK-STAT, MAPK, and PI3K pathways.

IL-15 has a wide range of biological activities, including promoting the survival and proliferation of T cells and NK cells, enhancing their cytotoxic activity, and regulating their differentiation and maturation. It also plays a role in the development and maintenance of memory T cells, which are critical for long-term immunity to pathogens.

Dysregulation of IL-15 signaling has been implicated in various diseases, including autoimmune disorders, chronic inflammation, and cancer. Therefore, IL-15 is a potential target for therapeutic intervention in these conditions.

"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.

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.

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.

MART-1, also known as Melanoma Antigen Recognized by T-Cells 1 or Melan-A, is a protein that is primarily found in melanocytes, which are the pigment-producing cells located in the skin, eyes, and hair follicles. It is a member of the family of antigens called melanoma differentiation antigens (MDAs) that are specifically expressed in melanocytes and melanomas. MART-1 is considered a tumor-specific antigen because it is overexpressed in melanoma cells compared to normal cells, making it an attractive target for immunotherapy.

MART-1 is presented on the surface of melanoma cells in complex with major histocompatibility complex (MHC) class I molecules, where it can be recognized by cytotoxic T lymphocytes (CTLs). This recognition triggers an immune response that can lead to the destruction of melanoma cells. MART-1 has been widely used as a target in various immunotherapy approaches, including cancer vaccines and adoptive cell transfer therapies, with the goal of enhancing the body's own immune system to recognize and eliminate melanoma cells.

A hapten is a small molecule that can elicit an immune response only when it is attached to a larger carrier protein. On its own, a hapten is too small to be recognized by the immune system as a foreign substance. However, when it binds to a carrier protein, it creates a new antigenic site that can be detected by the immune system. This process is known as haptenization.

Haptens are important in the study of immunology and allergies because they can cause an allergic response when they bind to proteins in the body. For example, certain chemicals found in cosmetics, drugs, or industrial products can act as haptens and trigger an allergic reaction when they come into contact with the skin or mucous membranes. The resulting immune response can cause symptoms such as rash, itching, or inflammation.

Haptens can also be used in the development of vaccines and diagnostic tests, where they are attached to carrier proteins to stimulate an immune response and produce specific antibodies that can be measured or used for therapy.

'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.

A binding site on an antibody refers to the specific region on the surface of the antibody molecule that can recognize and bind to a specific antigen. Antibodies are proteins produced by the immune system in response to the presence of foreign substances called antigens. They have two main functions: to neutralize the harmful effects of antigens and to help eliminate them from the body.

The binding site of an antibody is located at the tips of its Y-shaped structure, formed by the variable regions of the heavy and light chains of the antibody molecule. These regions contain unique amino acid sequences that determine the specificity of the antibody for a particular antigen. The binding site can recognize and bind to a specific epitope or region on the antigen, forming an antigen-antibody complex.

The binding between the antibody and antigen is highly specific and depends on non-covalent interactions such as hydrogen bonds, van der Waals forces, and electrostatic attractions. This interaction plays a crucial role in the immune response, as it allows the immune system to recognize and eliminate pathogens and other foreign substances from the body.

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.

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.

CCR7 (C-C chemokine receptor type 7) is a type of protein found on the surface of certain immune cells, including T cells and dendritic cells. It is a type of G protein-coupled receptor that binds to specific chemokines, which are small signaling proteins that help regulate the migration and activation of immune cells during an immune response.

CCR7 recognizes and binds to two main chemokines, CCL19 and CCL21, which are produced by specialized cells in lymphoid organs such as lymph nodes and the spleen. When CCR7 on an immune cell binds to one of these chemokines, it triggers a series of intracellular signaling events that cause the cell to migrate towards the source of the chemokine.

This process is important for the proper functioning of the immune system, as it helps to coordinate the movement of immune cells between different tissues and organs during an immune response. For example, dendritic cells in the peripheral tissues can use CCR7 to migrate to the draining lymph nodes, where they can present antigens to T cells and help stimulate an adaptive immune response. Similarly, activated T cells can use CCR7 to migrate to the site of an infection or inflammation, where they can carry out their effector functions.

I'm sorry for any confusion, but "Hemolytic Plaque Technique" is not a widely recognized or established medical term. It seems like it might be a combination of two different concepts in medical and scientific research: the Hemolytic Assay and the Plaque Assay technique.

A Hemolytic Assay is a method used to measure the amount of hemolysis, or the rupturing of red blood cells, caused by a substance such as a toxin or an antibody. This assay can help determine the concentration of the hemolysin in a sample.

On the other hand, the Plaque Assay Technique is a method used to measure the number of infectious virus particles in a sample. It involves adding a layer of cells (like bacteria) that the virus can infect and then covering it with a nutrient agar overlay. After a period of incubation, clear areas or "plaques" appear in the agar where the viruses have infected and lysed the cells. By counting these plaques, researchers can estimate the number of infectious virus particles present in the original sample.

Therefore, if you're looking for a definition of a Hemolytic Plaque Technique, it might refer to a research method that combines both concepts, possibly measuring the amount of a substance (like an antibody) that causes hemolysis in red blood cells and correlating it with the number of infectious virus particles present. However, I would recommend consulting the original source or author for clarification on their intended meaning.

Thy-1, also known as Thy-1 antigen or CD90, is a glycosylphosphatidylinositol (GPI)-anchored protein found on the surface of various cells in the body. It was first discovered as a cell surface antigen on thymocytes, hence the name Thy-1.

Thy-1 is a member of the immunoglobulin superfamily and is widely expressed in different tissues, including the brain, where it is found on the surface of neurons and glial cells. In the immune system, Thy-1 is expressed on the surface of T lymphocytes, natural killer (NK) cells, and some subsets of dendritic cells.

The function of Thy-1 is not fully understood, but it has been implicated in various biological processes, including cell adhesion, signal transduction, and regulation of immune responses. Thy-1 has also been shown to play a role in the development and maintenance of the nervous system, as well as in the pathogenesis of certain neurological disorders.

As an antigen, Thy-1 can be recognized by specific antibodies, which can be used in various research and clinical applications, such as immunohistochemistry, flow cytometry, and cell sorting.

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.

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.

CXCR3 is a type of chemokine receptor that is primarily expressed on the surface of certain immune cells, including T lymphocytes (a type of white blood cell involved in immune response). It belongs to the Class A orphan G protein-coupled receptors family.

CXCR3 has three known subtypes, CXCR3-A, CXCR3-B, and CXCR3-C, each with different roles in regulating immune cell functions. These receptors bind to specific chemokines, which are small signaling proteins that help direct the movement of immune cells towards sites of inflammation or infection.

The chemokines that bind to CXCR3 include CXCL9, CXCL10, and CXCL11, which are produced by various cell types in response to inflammation or injury. Once bound to these chemokines, CXCR3 activates intracellular signaling pathways that trigger a range of responses, such as cell migration, activation, and proliferation.

In the context of disease, CXCR3 has been implicated in various pathological conditions, including cancer, autoimmune diseases, and viral infections, due to its role in regulating immune cell trafficking and activation.

Immunodominant epitopes refer to specific regions or segments on an antigen (a molecule that can trigger an immune response) that are particularly effective at stimulating an immune response. These epitopes are often the parts of the antigen that are most recognized by the immune system, and as a result, they elicit a strong response from immune cells such as T-cells or B-cells.

In the context of T-cell responses, immunodominant epitopes are typically short peptide sequences (usually 8-15 amino acids long) that are presented to T-cells by major histocompatibility complex (MHC) molecules on the surface of antigen-presenting cells. The T-cell receptor recognizes and binds to these epitopes, triggering a cascade of immune responses aimed at eliminating the pathogen or foreign substance that contains the antigen.

In some cases, immunodominant epitopes may be the primary targets of vaccines or other immunotherapies, as they can elicit strong and protective immune responses. However, in other cases, immunodominant epitopes may also be associated with immune evasion or tolerance, where the immune system fails to mount an effective response against a pathogen or cancer cell. Understanding the properties and behavior of immunodominant epitopes is therefore crucial for developing effective vaccines and immunotherapies.

HLA-A1 antigen is a type of human leukocyte antigen (HLA) class I molecule that plays an important role in the immune system. The HLAs are proteins found on the surface of cells that help the immune system distinguish between the body's own cells and foreign substances, such as viruses and bacteria.

The HLA-A1 antigen is one of several different types of HLA-A molecules, and it is determined by a specific set of genes located on chromosome 6. The HLA-A1 antigen is expressed on the surface of some cells in the human body and can be detected through laboratory testing.

The HLA-A1 antigen is associated with certain diseases or conditions, such as an increased risk of developing certain types of cancer or autoimmune disorders. It is also used as a marker for tissue typing in organ transplantation to help match donors and recipients and reduce the risk of rejection.

It's important to note that the presence or absence of HLA-A1 antigen alone does not determine whether someone will develop a particular disease or experience a successful organ transplant. Other genetic and environmental factors also play a role in these outcomes.

Genetic transduction is a process in molecular biology that describes the transfer of genetic material from one bacterium to another by a viral vector called a bacteriophage (or phage). In this process, the phage infects one bacterium and incorporates a portion of the bacterial DNA into its own genetic material. When the phage then infects a second bacterium, it can transfer the incorporated bacterial DNA to the new host. This can result in the horizontal gene transfer (HGT) of traits such as antibiotic resistance or virulence factors between bacteria.

There are two main types of transduction: generalized and specialized. In generalized transduction, any portion of the bacterial genome can be packaged into the phage particle, leading to a random assortment of genetic material being transferred. In specialized transduction, only specific genes near the site where the phage integrates into the bacterial chromosome are consistently transferred.

It's important to note that genetic transduction is not to be confused with transformation or conjugation, which are other mechanisms of HGT in bacteria.

"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.

"Age factors" refer to the effects, changes, or differences that age can have on various aspects of health, disease, and medical care. These factors can encompass a wide range of issues, including:

1. Physiological changes: As people age, their bodies undergo numerous physical changes that can affect how they respond to medications, illnesses, and medical procedures. For example, older adults may be more sensitive to certain drugs or have weaker immune systems, making them more susceptible to infections.
2. Chronic conditions: Age is a significant risk factor for many chronic diseases, such as heart disease, diabetes, cancer, and arthritis. As a result, age-related medical issues are common and can impact treatment decisions and outcomes.
3. Cognitive decline: Aging can also lead to cognitive changes, including memory loss and decreased decision-making abilities. These changes can affect a person's ability to understand and comply with medical instructions, leading to potential complications in their care.
4. Functional limitations: Older adults may experience physical limitations that impact their mobility, strength, and balance, increasing the risk of falls and other injuries. These limitations can also make it more challenging for them to perform daily activities, such as bathing, dressing, or cooking.
5. Social determinants: Age-related factors, such as social isolation, poverty, and lack of access to transportation, can impact a person's ability to obtain necessary medical care and affect their overall health outcomes.

Understanding age factors is critical for healthcare providers to deliver high-quality, patient-centered care that addresses the unique needs and challenges of older adults. By taking these factors into account, healthcare providers can develop personalized treatment plans that consider a person's age, physical condition, cognitive abilities, and social circumstances.

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.

Autoimmunity is a medical condition in which the body's immune system mistakenly attacks and destroys healthy tissues within the body. In normal function, the immune system recognizes and fights off foreign substances such as bacteria, viruses, and toxins. However, when autoimmunity occurs, the immune system identifies self-molecules or tissues as foreign and produces an immune response against them.

This misguided response can lead to chronic inflammation, tissue damage, and impaired organ function. Autoimmune diseases can affect various parts of the body, including the joints, skin, glands, muscles, and blood vessels. Some common examples of autoimmune diseases are rheumatoid arthritis, lupus, multiple sclerosis, type 1 diabetes, Hashimoto's thyroiditis, and Graves' disease.

The exact cause of autoimmunity is not fully understood, but it is believed to involve a combination of genetic, environmental, and lifestyle factors that trigger an abnormal immune response in susceptible individuals. Treatment for autoimmune diseases typically involves managing symptoms, reducing inflammation, and suppressing the immune system's overactive response using medications such as corticosteroids, immunosuppressants, and biologics.

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.

Prognosis is a medical term that refers to the prediction of the likely outcome or course of a disease, including the chances of recovery or recurrence, based on the patient's symptoms, medical history, physical examination, and diagnostic tests. It is an important aspect of clinical decision-making and patient communication, as it helps doctors and patients make informed decisions about treatment options, set realistic expectations, and plan for future care.

Prognosis can be expressed in various ways, such as percentages, categories (e.g., good, fair, poor), or survival rates, depending on the nature of the disease and the available evidence. However, it is important to note that prognosis is not an exact science and may vary depending on individual factors, such as age, overall health status, and response to treatment. Therefore, it should be used as a guide rather than a definitive forecast.

A neoplasm is a tumor or growth that is formed by an abnormal and excessive proliferation of cells, which can be benign or malignant. Neoplasm proteins are therefore any proteins that are expressed or produced in these neoplastic cells. These proteins can play various roles in the development, progression, and maintenance of neoplasms.

Some neoplasm proteins may contribute to the uncontrolled cell growth and division seen in cancer, such as oncogenic proteins that promote cell cycle progression or inhibit apoptosis (programmed cell death). Others may help the neoplastic cells evade the immune system, allowing them to proliferate undetected. Still others may be involved in angiogenesis, the formation of new blood vessels that supply the tumor with nutrients and oxygen.

Neoplasm proteins can also serve as biomarkers for cancer diagnosis, prognosis, or treatment response. For example, the presence or level of certain neoplasm proteins in biological samples such as blood or tissue may indicate the presence of a specific type of cancer, help predict the likelihood of cancer recurrence, or suggest whether a particular therapy will be effective.

Overall, understanding the roles and behaviors of neoplasm proteins can provide valuable insights into the biology of cancer and inform the development of new diagnostic and therapeutic strategies.

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.

A hybridoma is a type of hybrid cell that is created in a laboratory by fusing a cancer cell (usually a B cell) with a normal immune cell. The resulting hybrid cell combines the ability of the cancer cell to grow and divide indefinitely with the ability of the immune cell to produce antibodies, which are proteins that help the body fight infection.

Hybridomas are commonly used to produce monoclonal antibodies, which are identical copies of a single antibody produced by a single clone of cells. These antibodies can be used for a variety of purposes, including diagnostic tests and treatments for diseases such as cancer and autoimmune disorders.

To create hybridomas, B cells are first isolated from the spleen or blood of an animal that has been immunized with a specific antigen (a substance that triggers an immune response). The B cells are then fused with cancer cells using a chemical agent such as polyethylene glycol. The resulting hybrid cells are called hybridomas and are grown in culture medium, where they can be selected for their ability to produce antibodies specific to the antigen of interest. These antibody-producing hybridomas can then be cloned to produce large quantities of monoclonal antibodies.

Anti-idiotypic antibodies are a type of immune protein that recognizes and binds to the unique identifying region (idiotype) of another antibody. These antibodies are produced by the immune system as part of a regulatory feedback mechanism, where they can modulate or inhibit the activity of the original antibody. They have been studied for their potential use in immunotherapy and vaccine development.

CD43, also known as leukosialin or sialophorin, is a protein found on the surface of various types of immune cells, including T cells, B cells, and natural killer (NK) cells. It is a type of transmembrane glycoprotein that is involved in cell-cell interactions, adhesion, and signaling.

CD43 is not typically considered an antigen in the traditional sense, as it does not elicit an immune response on its own. However, it can be used as a marker for identifying certain types of cells, particularly those of hematopoietic origin (i.e., cells that give rise to blood cells).

CD43 is also a target for some immunotherapy approaches, such as monoclonal antibody therapy, in the treatment of certain types of cancer. By binding to CD43 on the surface of cancer cells, these therapies aim to trigger an immune response against the cancer cells and promote their destruction.

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.

HIV (Human Immunodeficiency Virus) is a species of lentivirus (a subgroup of retrovirus) that causes HIV infection and over time, HIV infection can lead to AIDS (Acquired Immunodeficiency Syndrome). This virus attacks the immune system, specifically the CD4 cells, also known as T cells, which are a type of white blood cell that helps coordinate the body's immune response. As HIV destroys these cells, the body becomes more vulnerable to other infections and diseases. It is primarily spread through bodily fluids like blood, semen, vaginal fluids, and breast milk.

It's important to note that while there is no cure for HIV, with proper medical care, HIV can be controlled. Treatment for HIV is called antiretroviral therapy (ART). If taken as prescribed, this medicine reduces the amount of HIV in the body to a very low level, which keeps the immune system working and prevents illness. This treatment also greatly reduces the risk of transmission.

Immunologic techniques are a group of laboratory methods that utilize the immune system's ability to recognize and respond to specific molecules, known as antigens. These techniques are widely used in medicine, biology, and research to detect, measure, or identify various substances, including proteins, hormones, viruses, bacteria, and other antigens.

Some common immunologic techniques include:

1. Enzyme-linked Immunosorbent Assay (ELISA): A sensitive assay used to detect and quantify antigens or antibodies in a sample. This technique uses an enzyme linked to an antibody or antigen, which reacts with a substrate to produce a colored product that can be measured and quantified.
2. Immunofluorescence: A microscopic technique used to visualize the location of antigens or antibodies in tissues or cells. This technique uses fluorescent dyes conjugated to antibodies, which bind to specific antigens and emit light when excited by a specific wavelength of light.
3. Western Blotting: A laboratory technique used to detect and identify specific proteins in a sample. This technique involves separating proteins based on their size using electrophoresis, transferring them to a membrane, and then probing the membrane with antibodies that recognize the protein of interest.
4. Immunoprecipitation: A laboratory technique used to isolate and purify specific antigens or antibodies from a complex mixture. This technique involves incubating the mixture with an antibody that recognizes the antigen or antibody of interest, followed by precipitation of the antigen-antibody complex using a variety of methods.
5. Radioimmunoassay (RIA): A sensitive assay used to detect and quantify antigens or antibodies in a sample. This technique uses radioactively labeled antigens or antibodies, which bind to specific antigens or antibodies in the sample, allowing for detection and quantification using a scintillation counter.

These techniques are important tools in medical diagnosis, research, and forensic science.

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.

A radiation chimera is not a widely used or recognized medical term. However, in the field of genetics and radiation biology, a "chimera" refers to an individual that contains cells with different genetic backgrounds. A radiation chimera, therefore, could refer to an organism that has become a chimera as a result of exposure to radiation, which can cause mutations and changes in the genetic makeup of cells.

Ionizing radiation, such as that used in cancer treatments or nuclear accidents, can cause DNA damage and mutations in cells. If an organism is exposed to radiation and some of its cells undergo mutations while others do not, this could result in a chimera with genetically distinct populations of cells.

However, it's important to note that the term "radiation chimera" is not commonly used in medical literature or clinical settings. If you encounter this term in a different context, I would recommend seeking clarification from the source to ensure a proper understanding.

Prospective studies, also known as longitudinal studies, are a type of cohort study in which data is collected forward in time, following a group of individuals who share a common characteristic or exposure over a period of time. The researchers clearly define the study population and exposure of interest at the beginning of the study and follow up with the participants to determine the outcomes that develop over time. This type of study design allows for the investigation of causal relationships between exposures and outcomes, as well as the identification of risk factors and the estimation of disease incidence rates. Prospective studies are particularly useful in epidemiology and medical research when studying diseases with long latency periods or rare outcomes.

Chemokine (C-C motif) ligand 21 (CCL21), also known as secondary lymphoid tissue chemokine (SLC) or exodus-2, is a type of chemokine that belongs to the CC subfamily. Chemokines are small signaling proteins that play crucial roles in regulating immune responses and inflammation by recruiting various leukocytes to sites of infection or injury through specific receptor binding.

CCL21 is primarily expressed in high endothelial venules (HEVs) within lymphoid tissues, such as lymph nodes, spleen, and Peyer's patches. It functions as a chemoattractant for immune cells like dendritic cells, T cells, and B cells, guiding them to enter the HEVs and migrate into the lymphoid organs. This process is essential for initiating adaptive immune responses against pathogens or antigens.

CCL21 exerts its effects by binding to chemokine receptors CCR7 and atypical chemokine receptor ACKR3 (also known as CXCR7). The interaction between CCL21 and these receptors triggers intracellular signaling cascades, leading to cell migration and activation. Dysregulation of CCL21 expression or function has been implicated in various pathological conditions, including autoimmune diseases, cancer, and inflammatory disorders.

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.

Vaccinia virus is a large, complex DNA virus that belongs to the Poxviridae family. It is the virus used in the production of the smallpox vaccine. The vaccinia virus is not identical to the variola virus, which causes smallpox, but it is closely related and provides cross-protection against smallpox infection.

The vaccinia virus has a unique replication cycle that occurs entirely in the cytoplasm of infected cells, rather than in the nucleus like many other DNA viruses. This allows the virus to evade host cell defenses and efficiently produce new virions. The virus causes the formation of pocks or lesions on the skin, which contain large numbers of virus particles that can be transmitted to others through close contact.

Vaccinia virus has also been used as a vector for the delivery of genes encoding therapeutic proteins, vaccines against other infectious diseases, and cancer therapies. However, the use of vaccinia virus as a vector is limited by its potential to cause adverse reactions in some individuals, particularly those with weakened immune systems or certain skin conditions.

Diabetes Mellitus, Type 1 is a chronic autoimmune disease characterized by the destruction of insulin-producing beta cells in the pancreas, leading to an absolute deficiency of insulin. This results in an inability to regulate blood glucose levels, causing hyperglycemia (high blood sugar). Type 1 diabetes typically presents in childhood or early adulthood, although it can develop at any age. It is usually managed with regular insulin injections or the use of an insulin pump, along with monitoring of blood glucose levels and adjustments to diet and physical activity. Uncontrolled type 1 diabetes can lead to serious complications such as kidney damage, nerve damage, blindness, and cardiovascular disease.

CD40 ligand (CD40L or CD154) is a type II transmembrane protein and a member of the tumor necrosis factor (TNF) superfamily. It is primarily expressed on activated CD4+ T cells, but can also be found on other immune cells such as activated B cells, macrophages, and dendritic cells.

CD40 ligand binds to its receptor, CD40, which is mainly expressed on the surface of antigen-presenting cells (APCs) such as B cells, dendritic cells, and macrophages. The interaction between CD40L and CD40 plays a crucial role in the activation and regulation of the immune response.

CD40L-CD40 signaling is essential for T cell-dependent B cell activation, antibody production, and class switching. It also contributes to the activation and maturation of dendritic cells, promoting their ability to stimulate T cell responses. Dysregulation of CD40L-CD40 signaling has been implicated in various autoimmune diseases, transplant rejection, and cancer.

Beta-chain gene rearrangement in the T-cell antigen receptor (TCR) refers to the genetic process that occurs during the development of T cells, a type of white blood cell crucial for adaptive immunity. The TCR is a heterodimeric protein complex expressed on the surface of T cells, responsible for recognizing and binding to specific peptide antigens presented in the context of major histocompatibility complex (MHC) molecules.

The beta-chain of the TCR is encoded by a set of gene segments called V (variable), D (diversity), J (joining), and C (constant) segments, located on chromosome 7 in humans. During T-cell development in the thymus, the following rearrangement events occur:

1. A random selection and recombination of a V, D, and J segment take place, forming a variable region exon that encodes the antigen-binding site of the beta-chain. This process introduces nucleotide insertions or deletions at the junctions between these segments, further increasing diversity.
2. The rearranged VDJ segment then combines with a C segment through RNA splicing to form a continuous mRNA sequence that encodes the complete beta-chain protein.
3. The resulting beta-chain pairs with an alpha-chain (encoded by similar gene segments on chromosome 14) to create a functional TCR heterodimer, which is then expressed on the T-cell surface.

This gene rearrangement process allows for the generation of a vast array of unique TCRs capable of recognizing various peptide antigens, ensuring broad coverage against potential pathogens and tumor cells.

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.

Simian Acquired Immunodeficiency Syndrome (SAIDS) is not recognized as a medical condition in humans. However, it is a disease that affects non-human primates like African green monkeys and sooty mangabeys. SAIDS is caused by the Simian Immunodeficiency Virus (SIV), which is similar to the Human Immunodeficiency Virus (HIV) that leads to Acquired Immunodeficiency Syndrome (AIDS) in humans.

In non-human primates, SIV infection can lead to a severe immunodeficiency state, characterized by the destruction of CD4+ T cells and impaired immune function, making the host susceptible to various opportunistic infections and cancers. However, it is important to note that most non-human primates infected with SIV do not develop SAIDS spontaneously, unlike humans who acquire HIV infection.

In summary, Simian Acquired Immunodeficiency Syndrome (SAIDS) is a disease affecting non-human primates due to Simian Immunodeficiency Virus (SIV) infection, characterized by immunodeficiency and susceptibility to opportunistic infections and cancers. It should not be confused with Human Immunodeficiency Virus Infection and Acquired Immunodeficiency Syndrome (HIV/AIDS) in humans.

Peyer's patches are specialized lymphoid nodules found in the mucosa of the ileum, a part of the small intestine. They are a component of the immune system and play a crucial role in monitoring and defending against harmful pathogens that are ingested with food and drink. Peyer's patches contain large numbers of B-lymphocytes, T-lymphocytes, and macrophages, which work together to identify and eliminate potential threats. They also have a unique structure that allows them to sample and analyze the contents of the intestinal lumen, providing an early warning system for the immune system.

Leukemia is a type of cancer that originates from the bone marrow - the soft, inner part of certain bones where new blood cells are made. It is characterized by an abnormal production of white blood cells, known as leukocytes or blasts. These abnormal cells accumulate in the bone marrow and interfere with the production of normal blood cells, leading to a decrease in red blood cells (anemia), platelets (thrombocytopenia), and healthy white blood cells (leukopenia).

There are several types of leukemia, classified based on the specific type of white blood cell affected and the speed at which the disease progresses:

1. Acute Leukemias - These types of leukemia progress rapidly, with symptoms developing over a few weeks or months. They involve the rapid growth and accumulation of immature, nonfunctional white blood cells (blasts) in the bone marrow and peripheral blood. The two main categories are:
- Acute Lymphoblastic Leukemia (ALL) - Originates from lymphoid progenitor cells, primarily affecting children but can also occur in adults.
- Acute Myeloid Leukemia (AML) - Develops from myeloid progenitor cells and is more common in older adults.

2. Chronic Leukemias - These types of leukemia progress slowly, with symptoms developing over a period of months to years. They involve the production of relatively mature, but still abnormal, white blood cells that can accumulate in large numbers in the bone marrow and peripheral blood. The two main categories are:
- Chronic Lymphocytic Leukemia (CLL) - Affects B-lymphocytes and is more common in older adults.
- Chronic Myeloid Leukemia (CML) - Originates from myeloid progenitor cells, characterized by the presence of a specific genetic abnormality called the Philadelphia chromosome. It can occur at any age but is more common in middle-aged and older adults.

Treatment options for leukemia depend on the type, stage, and individual patient factors. Treatments may include chemotherapy, targeted therapy, immunotherapy, stem cell transplantation, or a combination of these approaches.

Lymphocytic choriomeningitis virus (LCMV) is an Old World arenavirus that primarily infects rodents, particularly the house mouse (Mus musculus). The virus is harbored in these mice without causing any apparent disease, but they can shed the virus in their urine, droppings, and saliva.

Humans can contract LCMV through close contact with infected rodents or their excreta, inhalation of aerosolized virus, or ingestion of contaminated food or water. In humans, LCMV infection can cause a mild to severe illness called lymphocytic choriomeningitis (LCM), which primarily affects the meninges (the membranes surrounding the brain and spinal cord) and, less frequently, the brain and spinal cord itself.

The incubation period for LCMV infection is typically 1-2 weeks, after which symptoms may appear. Initial symptoms include fever, malaise, headache, muscle aches, and nausea. In some cases, the illness may progress to involve the meninges (meningitis), resulting in neck stiffness, light sensitivity, and altered mental status. In rare instances, LCMV infection can lead to encephalitis (inflammation of the brain) or myelitis (inflammation of the spinal cord), causing more severe neurological symptoms such as seizures, paralysis, or long-term neurological damage.

Most individuals who contract LCMV recover completely within a few weeks to months; however, immunocompromised individuals are at risk for developing severe and potentially fatal complications from the infection. Pregnant women infected with LCMV may also face an increased risk of miscarriage or fetal abnormalities.

Prevention measures include avoiding contact with rodents, especially house mice, and their excreta, maintaining good hygiene, and using appropriate personal protective equipment when handling potentially contaminated materials. There is no specific treatment for LCMV infection; management typically involves supportive care to alleviate symptoms and address complications as they arise.

Major Histocompatibility Complex (MHC) class I genes are a group of genes that encode proteins found on the surface of most nucleated cells in the body. These proteins play a crucial role in the immune system by presenting pieces of protein from inside the cell to T-cells, which are a type of white blood cell. This process allows the immune system to detect and respond to cells that have been infected by viruses or become cancerous.

MHC class I genes are highly polymorphic, meaning there are many different variations of these genes in the population. This diversity is important for the immune system's ability to recognize and respond to a wide variety of pathogens. The MHC class I proteins are composed of three main regions: the heavy chain, which is encoded by the MHC class I gene; a short peptide, which is derived from inside the cell; and a light chain called beta-2 microglobulin, which is not encoded by an MHC gene.

There are three major types of MHC class I genes in humans, known as HLA-A, HLA-B, and HLA-C. These genes are located on chromosome 6 and are among the most polymorphic genes in the human genome. The products of these genes are critical for the immune system's ability to distinguish between self and non-self, and play a key role in organ transplant rejection.

Antigens are substances that can stimulate an immune response, particularly the production of antibodies by B-lymphocytes. Differentiation refers to the process by which cells mature and become more specialized in their functions. In the context of B-lymphocytes, differentiation involves the maturation of naive B-cells into plasma cells that are capable of producing large amounts of antibodies in response to an antigenic stimulus.

B-lymphocytes, also known as B-cells, are a type of white blood cell that plays a critical role in the adaptive immune system. They are responsible for producing antibodies, which are proteins that recognize and bind to specific antigens, marking them for destruction by other immune cells.

When a B-lymphocyte encounters an antigen, it becomes activated and begins to differentiate into a plasma cell. During this process, the B-cell undergoes several changes, including an increase in size, the expression of new surface receptors, and the production of large amounts of antibodies specific to the antigen. These antibodies are then released into the bloodstream, where they can bind to the antigen and help to neutralize or eliminate it.

Overall, the differentiation of B-lymphocytes in response to antigens is a critical component of the adaptive immune system, allowing the body to mount targeted responses to specific pathogens and other foreign substances.

Anti-HIV agents are a class of medications specifically designed to treat HIV (Human Immunodeficiency Virus) infection. These drugs work by interfering with various stages of the HIV replication cycle, preventing the virus from infecting and killing CD4+ T cells, which are crucial for maintaining a healthy immune system.

There are several classes of anti-HIV agents, including:

1. Nucleoside/Nucleotide Reverse Transcriptase Inhibitors (NRTIs): These drugs act as faulty building blocks that the virus incorporates into its genetic material, causing the replication process to halt. Examples include zidovudine (AZT), lamivudine (3TC), and tenofovir.
2. Non-nucleoside Reverse Transcriptase Inhibitors (NNRTIs): These medications bind directly to the reverse transcriptase enzyme, altering its shape and preventing it from functioning properly. Examples include efavirenz, nevirapine, and rilpivirine.
3. Protease Inhibitors (PIs): These drugs target the protease enzyme, which is responsible for cleaving viral polyproteins into functional components. By inhibiting this enzyme, PIs prevent the formation of mature, infectious virus particles. Examples include atazanavir, darunavir, and lopinavir.
4. Integrase Strand Transfer Inhibitors (INSTIs): These medications block the integrase enzyme, which is responsible for inserting the viral genetic material into the host cell's DNA. By inhibiting this step, INSTIs prevent the virus from establishing a permanent infection within the host cell. Examples include raltegravir, dolutegravir, and bictegravir.
5. Fusion/Entry Inhibitors: These drugs target different steps of the viral entry process, preventing HIV from infecting CD4+ T cells. Examples include enfuvirtide (T-20), maraviroc, and ibalizumab.
6. Post-Attachment Inhibitors: This class of medications prevents the virus from attaching to the host cell's receptors, thereby inhibiting infection. Currently, there is only one approved post-attachment inhibitor, fostemsavir.

Combination therapy using multiple classes of antiretroviral drugs has been shown to effectively suppress viral replication and improve clinical outcomes in people living with HIV. Regular adherence to the prescribed treatment regimen is crucial for maintaining an undetectable viral load and reducing the risk of transmission.

Lymphokine-activated killer (LAK) cells are a type of immune cell that has been activated to kill certain types of cells, including cancer cells and virus-infected cells. They are called "lymphokine-activated" because they are activated through the action of lymphokines, which are proteins secreted by other immune cells. LAK cells are a type of natural killer (NK) cell, which are a type of white blood cell that plays a role in the body's defense against viruses and cancer.

LAK cells are generated in the laboratory by incubating peripheral blood mononuclear cells (PBMCs), which include lymphocytes and monocytes, with high concentrations of interleukin-2 (IL-2) for several days. This process activates and expands the population of NK cells, resulting in the formation of LAK cells. These activated cells are then able to recognize and kill a wide range of tumor cells and virus-infected cells, regardless of whether they express specific antigens or not.

LAK cell therapy is an experimental form of cancer treatment that involves infusing patients with large numbers of LAK cells in order to enhance their immune response against cancer. While some studies have shown promising results, more research is needed to determine the safety and effectiveness of this approach.

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.

Major Histocompatibility Complex (MHC) Class II genes are a group of genes that encode cell surface proteins responsible for presenting peptide antigens to CD4+ T cells, which are crucial in the adaptive immune response. These proteins are expressed mainly on professional antigen-presenting cells such as dendritic cells, macrophages, and B cells. MHC Class II molecules present extracellular antigens derived from bacteria, viruses, and other pathogens, facilitating the activation of appropriate immune responses to eliminate the threat. The genes responsible for these proteins are found within the MHC locus on chromosome 6 in humans (chromosome 17 in mice).

Mucosal immunity refers to the immune system's defense mechanisms that are specifically adapted to protect the mucous membranes, which line various body openings such as the respiratory, gastrointestinal, and urogenital tracts. These membranes are constantly exposed to foreign substances, including potential pathogens, and therefore require a specialized immune response to maintain homeostasis and prevent infection.

Mucosal immunity is primarily mediated by secretory IgA (SIgA) antibodies, which are produced by B cells in the mucosa-associated lymphoid tissue (MALT). These antibodies can neutralize pathogens and prevent them from adhering to and invading the epithelial cells that line the mucous membranes.

In addition to SIgA, other components of the mucosal immune system include innate immune cells such as macrophages, dendritic cells, and neutrophils, which can recognize and respond to pathogens through pattern recognition receptors (PRRs). T cells also play a role in mucosal immunity, particularly in the induction of cell-mediated immunity against viruses and other intracellular pathogens.

Overall, mucosal immunity is an essential component of the body's defense system, providing protection against a wide range of potential pathogens while maintaining tolerance to harmless antigens present in the environment.

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.

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.

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.

'Inbred AKR mice' is a strain of laboratory mice used in biomedical research. The 'AKR' designation stands for "Akita Radioactive," referring to the location where this strain was first developed in Akita, Japan. These mice are inbred, meaning that they have been produced by many generations of brother-sister matings, resulting in a genetically homogeneous population with minimal genetic variation.

Inbred AKR mice are known for their susceptibility to certain types of leukemia and lymphoma, making them valuable models for studying these diseases and testing potential therapies. They also develop age-related cataracts and have a higher incidence of diabetes than some other strains.

It is important to note that while inbred AKR mice are widely used in research, their genetic uniformity may limit the applicability of findings to more genetically diverse human populations.

Serine endopeptidases are a type of enzymes that cleave peptide bonds within proteins (endopeptidases) and utilize serine as the nucleophilic amino acid in their active site for catalysis. These enzymes play crucial roles in various biological processes, including digestion, blood coagulation, and programmed cell death (apoptosis). Examples of serine endopeptidases include trypsin, chymotrypsin, thrombin, and elastase.

Nutrition disorders refer to conditions that result from eating, drinking, or absorbing nutrients in a way that is not consistent with human physiological needs. These disorders can manifest as both undernutrition and overnutrition. Undernutrition includes disorders such as protein-energy malnutrition, vitamin deficiencies, and mineral deficiencies, while overnutrition includes conditions such as obesity and diet-related noncommunicable diseases like diabetes, cardiovascular disease, and certain types of cancer.

Malnutrition is the broad term used to describe a state in which a person's nutrient intake is insufficient or excessive, leading to negative consequences for their health. Malnutrition can be caused by a variety of factors, including poverty, food insecurity, lack of education, cultural practices, and chronic diseases.

In addition to under- and overnutrition, disordered eating patterns such as anorexia nervosa, bulimia nervosa, binge eating disorder, and other specified feeding or eating disorders can also be considered nutrition disorders. These conditions are characterized by abnormal eating habits that can lead to serious health consequences, including malnutrition, organ damage, and mental health problems.

Overall, nutrition disorders are complex conditions that can have significant impacts on a person's physical and mental health. They require careful assessment, diagnosis, and treatment by healthcare professionals with expertise in nutrition and dietetics.

Autoantigens are substances that are typically found in an individual's own body, but can stimulate an immune response because they are recognized as foreign by the body's own immune system. In autoimmune diseases, the immune system mistakenly attacks and damages healthy tissues and organs because it recognizes some of their components as autoantigens. These autoantigens can be proteins, DNA, or other molecules that are normally present in the body but have become altered or exposed due to various factors such as infection, genetics, or environmental triggers. The immune system then produces antibodies and activates immune cells to attack these autoantigens, leading to tissue damage and inflammation.

Lymphoproliferative disorders (LPDs) are a group of diseases characterized by the excessive proliferation of lymphoid cells, which are crucial components of the immune system. These disorders can arise from both B-cells and T-cells, leading to various clinical manifestations ranging from benign to malignant conditions.

LPDs can be broadly classified into reactive and neoplastic categories:

1. Reactive Lymphoproliferative Disorders: These are typically triggered by infections, autoimmune diseases, or immunodeficiency states. They involve an exaggerated response of the immune system leading to the excessive proliferation of lymphoid cells. Examples include:
* Infectious mononucleosis (IM) caused by Epstein-Barr virus (EBV)
* Lymph node enlargement due to various infections or autoimmune disorders
* Post-transplant lymphoproliferative disorder (PTLD), which occurs in the context of immunosuppression following organ transplantation
2. Neoplastic Lymphoproliferative Disorders: These are malignant conditions characterized by uncontrolled growth and accumulation of abnormal lymphoid cells, leading to the formation of tumors. They can be further classified into Hodgkin lymphoma (HL) and non-Hodgkin lymphoma (NHL). Examples include:
* Hodgkin lymphoma (HL): Classical HL and nodular lymphocyte-predominant HL
* Non-Hodgkin lymphoma (NHL): Various subtypes, such as diffuse large B-cell lymphoma, follicular lymphoma, mantle cell lymphoma, and Burkitt lymphoma

It is important to note that the distinction between reactive and neoplastic LPDs can sometimes be challenging, requiring careful clinical, histopathological, immunophenotypic, and molecular evaluations. Proper diagnosis and classification of LPDs are crucial for determining appropriate treatment strategies and predicting patient outcomes.

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.

Cell transformation, viral refers to the process by which a virus causes normal cells to become cancerous or tumorigenic. This occurs when the genetic material of the virus integrates into the DNA of the host cell and alters its regulation, leading to uncontrolled cell growth and division. Some viruses known to cause cell transformation include human papillomavirus (HPV), hepatitis B virus (HBV), and certain types of herpesviruses.

Agammaglobulinemia is a medical condition characterized by a severe deficiency or complete absence of gamma globulins (a type of antibodies) in the blood. This deficiency results from a lack of functional B cells, which are a type of white blood cell that produces antibodies to help fight off infections.

There are two main types of agammaglobulinemia: X-linked agammaglobulinemia (XLA) and autosomal recessive agammaglobulinemia (ARA). XLA is caused by mutations in the BTK gene and primarily affects males, while ARA is caused by mutations in other genes and can affect both males and females.

People with agammaglobulinemia are at increased risk for recurrent bacterial infections, particularly respiratory tract infections such as pneumonia and sinusitis. They may also be more susceptible to certain viral and parasitic infections. Treatment typically involves replacement therapy with intravenous immunoglobulin (IVIG) to provide the patient with functional antibodies.

Trinitrobenzenes are not typically referred to in the context of medical definitions, as they are chemical compounds that do not have direct relevance to medical fields. However, I can provide you with some general information about trinitrobenzenes.

Trinitrobenzenes are a group of chemical compounds consisting of a benzene ring (a cyclic aromatic hydrocarbon) with three nitro groups (-NO2) attached to it. The most common and well-known compound in this group is 1,3,5-trinitrobenzene (TNB). These compounds are highly explosive and have been used in military and industrial applications for their energetic properties.

In a medical or clinical context, trinitrobenzenes may be encountered only if someone is exposed to these chemicals in an occupational or accidental setting. Exposure can lead to local skin irritation, eye damage, or respiratory issues. Chronic exposure or high-dose acute exposure might cause more severe health problems, including damage to the liver and kidneys. However, trinitrobenzenes are not used as therapeutic agents or diagnostic tools in medicine.

Nuclear factor of activated T-cells (NFAT) transcription factors are a group of proteins that play a crucial role in the regulation of gene transcription in various cells, including immune cells. They are involved in the activation of genes responsible for immune responses, cell survival, differentiation, and development.

NFAT transcription factors can be divided into five main members: NFATC1 (also known as NFAT2 or NFATp), NFATC2 (or NFAT1), NFATC3 (or NFATc), NFATC4 (or NFAT3), and NFAT5 (or TonEBP). These proteins share a highly conserved DNA-binding domain, known as the Rel homology region, which allows them to bind to specific sequences in the promoter or enhancer regions of target genes.

NFATC transcription factors are primarily located in the cytoplasm in their inactive form, bound to inhibitory proteins. Upon stimulation of the cell, typically through calcium-dependent signaling pathways, NFAT proteins get dephosphorylated by calcineurin phosphatase, leading to their nuclear translocation and activation. Once in the nucleus, NFATC transcription factors can form homodimers or heterodimers with other transcription factors, such as AP-1, to regulate gene expression.

In summary, NFATC transcription factors are a family of proteins involved in the regulation of gene transcription, primarily in immune cells, and play critical roles in various cellular processes, including immune responses, differentiation, and development.

"Gene products, GAG" refer to the proteins that are produced by the GAG (Group-specific Antigen) gene found in retroviruses, such as HIV (Human Immunodeficiency Virus). These proteins play a crucial role in the structure and function of the viral particle or virion.

The GAG gene encodes for a polyprotein that is cleaved by a protease into several individual proteins, including matrix (MA), capsid (CA), and nucleocapsid (NC) proteins. These proteins are involved in the formation of the viral core, which encloses the viral RNA genome and associated enzymes required for replication.

The MA protein is responsible for binding to the host cell membrane during viral entry, while the CA protein forms the capsid shell that surrounds the viral RNA and NC protein. The NC protein binds to the viral RNA and helps to package it into the virion during assembly. Overall, GAG gene products are essential for the life cycle of retroviruses and are important targets for antiretroviral therapy in HIV-infected individuals.

T-cell lymphoma is a type of cancer that affects the T-cells, which are a specific type of white blood cell responsible for immune function. These lymphomas develop from mature T-cells and can be classified into various subtypes based on their clinical and pathological features.

T-cell lymphomas can arise in many different organs, including the lymph nodes, skin, and other soft tissues. They often present with symptoms such as enlarged lymph nodes, fever, night sweats, and weight loss. The diagnosis of T-cell lymphoma typically involves a biopsy of the affected tissue, followed by immunophenotyping and genetic analysis to determine the specific subtype.

Treatment for T-cell lymphomas may include chemotherapy, radiation therapy, immunotherapy, or stem cell transplantation, depending on the stage and aggressiveness of the disease. The prognosis for T-cell lymphoma varies widely depending on the subtype and individual patient factors.

I could not find a specific medical definition for "Vaccines, DNA." However, I can provide you with some information about DNA vaccines.

DNA vaccines are a type of vaccine that uses genetically engineered DNA to stimulate an immune response in the body. They work by introducing a small piece of DNA into the body that contains the genetic code for a specific antigen (a substance that triggers an immune response). The cells of the body then use this DNA to produce the antigen, which prompts the immune system to recognize and attack it.

DNA vaccines have several advantages over traditional vaccines. They are relatively easy to produce, can be stored at room temperature, and can be designed to protect against a wide range of diseases. Additionally, because they use DNA to stimulate an immune response, DNA vaccines do not require the growth and culture of viruses or bacteria, which can make them safer than traditional vaccines.

DNA vaccines are still in the experimental stages, and more research is needed to determine their safety and effectiveness. However, they have shown promise in animal studies and are being investigated as a potential tool for preventing a variety of infectious diseases, including influenza, HIV, and cancer.

The H-Y antigen is a complex of historically significant, male-specific proteins that are encoded by genes on the Y chromosome. These antigens were first discovered through studies of tissue rejection in animal models and were later found to be important in the field of transplantation immunology.

In a medical definition, the H-Y antigen refers to a group of antigens that are expressed on the cell surface of nucleated cells in males, including those found in tissues such as skin, muscle, and blood cells. They are recognized by the immune system as foreign when transplanted into females, leading to a rejection response.

The H-Y antigen has been the subject of extensive research due to its role in sex determination and differentiation, as well as its potential implications for autoimmune diseases and cancer biology. However, it's worth noting that the clinical relevance of the H-Y antigen is limited, and its study is primarily of academic interest.

The Kv1.3 potassium channel is a type of voltage-gated potassium channel that is widely expressed in various tissues, including immune cells such as T lymphocytes. It plays a crucial role in regulating the electrical activity of cells and controlling the flow of potassium ions across the cell membrane.

Kv1.3 channels are composed of four pore-forming alpha subunits, each containing six transmembrane domains. These channels open and close in response to changes in the membrane potential, allowing potassium ions to flow out of the cell when the channel is open. This movement of ions helps to restore the resting membrane potential and regulate the excitability of the cell.

In T lymphocytes, Kv1.3 channels are involved in the regulation of calcium signaling and activation of immune responses. They play a critical role in maintaining the membrane potential and controlling the release of calcium from intracellular stores, which is necessary for T-cell activation and proliferation. Inhibition or blockade of Kv1.3 channels has been shown to suppress T-cell activation and could have potential therapeutic implications in the treatment of autoimmune diseases and transplant rejection.

Integrins are a type of cell-adhesion molecule that play a crucial role in cell-cell and cell-extracellular matrix (ECM) interactions. They are heterodimeric transmembrane receptors composed of non-covalently associated α and β subunits, which form more than 24 distinct integrin heterodimers in humans.

Integrins bind to specific ligands, such as ECM proteins (e.g., collagen, fibronectin, laminin), cell surface molecules, and soluble factors, through their extracellular domains. The intracellular domains of integrins interact with the cytoskeleton and various signaling proteins, allowing them to transduce signals from the ECM into the cell (outside-in signaling) and vice versa (inside-out signaling).

These molecular interactions are essential for numerous biological processes, including cell adhesion, migration, proliferation, differentiation, survival, and angiogenesis. Dysregulation of integrin function has been implicated in various pathological conditions, such as cancer, fibrosis, inflammation, and autoimmune diseases.

T-cell antigen receptor (TCR) specificity refers to the ability of a T-cell's antigen receptor to recognize and bind to a specific antigenic peptide presented in the context of a major histocompatibility complex (MHC) molecule on the surface of an antigen-presenting cell. The TCR is a protein complex found on the surface of T-cells, which plays a critical role in adaptive immunity by identifying and responding to infected or cancerous cells.

The specificity of the TCR is determined by the complementarity-determining regions (CDRs) within its variable domains. These CDRs form a binding site that recognizes and interacts with a specific epitope, typically an 8-12 amino acid long peptide, presented in the groove of an MHC molecule. The TCR-antigen interaction is highly specific, allowing T-cells to distinguish between self and non-self antigens and initiate an appropriate immune response.

In summary, T-cell antigen receptor specificity refers to the unique ability of a T-cell's antigen receptor to recognize and bind to a specific antigenic peptide presented in the context of an MHC molecule, which is critical for the initiation and regulation of adaptive immune responses.

Plasma cells are a type of white blood cell that are derived from B cells (another type of white blood cell) and are responsible for producing antibodies. Antibodies are proteins that help the body to fight against infections by recognizing and binding to specific antigens, such as bacteria or viruses. Plasma cells are found in the bone marrow, spleen, and lymph nodes, and they play a crucial role in the immune system's response to infection.

Plasma cells are characterized by their large size, eccentric nucleus, and abundant cytoplasm filled with rough endoplasmic reticulum, which is where antibody proteins are synthesized and stored. When activated, plasma cells can produce and secrete large amounts of antibodies into the bloodstream and lymphatic system, where they can help to neutralize or eliminate pathogens.

It's worth noting that while plasma cells play an important role in the immune response, abnormal accumulations of these cells can also be a sign of certain diseases, such as multiple myeloma, a type of cancer that affects plasma cells.

Neoplasms are abnormal growths of cells or tissues in the body that serve no physiological function. They can be benign (non-cancerous) or malignant (cancerous). Benign neoplasms are typically slow growing and do not spread to other parts of the body, while malignant neoplasms are aggressive, invasive, and can metastasize to distant sites.

Neoplasms occur when there is a dysregulation in the normal process of cell division and differentiation, leading to uncontrolled growth and accumulation of cells. This can result from genetic mutations or other factors such as viral infections, environmental exposures, or hormonal imbalances.

Neoplasms can develop in any organ or tissue of the body and can cause various symptoms depending on their size, location, and type. Treatment options for neoplasms include surgery, radiation therapy, chemotherapy, immunotherapy, and targeted therapy, among others.

Organ size refers to the volume or physical measurement of an organ in the body of an individual. It can be described in terms of length, width, and height or by using specialized techniques such as imaging studies (like CT scans or MRIs) to determine the volume. The size of an organ can vary depending on factors such as age, sex, body size, and overall health status. Changes in organ size may indicate various medical conditions, including growths, inflammation, or atrophy.

"T-lymphocyte gene rearrangement" refers to the process that occurs during the development of T-cells (a type of white blood cell) in which the genes that code for their antigen receptors are rearranged to create a unique receptor that can recognize and bind to specific foreign molecules, such as viruses or tumor cells.

The T-cell receptor (TCR) is made up of two chains, alpha and beta, which are composed of variable and constant regions. During gene rearrangement, the variable region genes are rearranged through a process called V(D)J recombination, in which specific segments of DNA are cut and joined together to form a unique combination that encodes for a diverse range of antigen receptors.

This allows T-cells to recognize and respond to a wide variety of foreign molecules, contributing to the adaptive immune response. However, this process can also lead to errors and the generation of T-cells with self-reactive receptors, which can contribute to autoimmune diseases if not properly regulated.

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.

Severe Combined Immunodeficiency (SCID) is a group of rare genetic disorders characterized by deficient or absent immune responses. It results from mutations in different genes involved in the development and function of T lymphocytes, B lymphocytes, or both, leading to a severe impairment in cell-mediated and humoral immunity.

Infants with SCID are extremely vulnerable to infections, which can be life-threatening. Common symptoms include chronic diarrhea, failure to thrive, recurrent pneumonia, and persistent candidiasis (thrush). If left untreated, it can lead to severe disability or death within the first two years of life. Treatment typically involves bone marrow transplantation or gene therapy to restore immune function.

Isoantibodies are antibodies produced by the immune system that recognize and react to antigens (markers) found on the cells or tissues of another individual of the same species. These antigens are typically proteins or carbohydrates present on the surface of red blood cells, but they can also be found on other cell types.

Isoantibodies are formed when an individual is exposed to foreign antigens, usually through blood transfusions, pregnancy, or tissue transplantation. The exposure triggers the immune system to produce specific antibodies against these antigens, which can cause a harmful immune response if the individual receives another transfusion or transplant from the same donor in the future.

There are two main types of isoantibodies:

1. Agglutinins: These are IgM antibodies that cause red blood cells to clump together (agglutinate) when mixed with the corresponding antigen. They develop rapidly after exposure and can cause immediate transfusion reactions or hemolytic disease of the newborn in pregnant women.
2. Hemolysins: These are IgG antibodies that destroy red blood cells by causing their membranes to become more permeable, leading to lysis (bursting) of the cells and release of hemoglobin into the plasma. They take longer to develop but can cause delayed transfusion reactions or hemolytic disease of the newborn in pregnant women.

Isoantibodies are detected through blood tests, such as the crossmatch test, which determines compatibility between a donor's and recipient's blood before transfusions or transplants.

"Cat" is a common name that refers to various species of small carnivorous mammals that belong to the family Felidae. The domestic cat, also known as Felis catus or Felis silvestris catus, is a popular pet and companion animal. It is a subspecies of the wildcat, which is found in Europe, Africa, and Asia.

Domestic cats are often kept as pets because of their companionship, playful behavior, and ability to hunt vermin. They are also valued for their ability to provide emotional support and therapy to people. Cats are obligate carnivores, which means that they require a diet that consists mainly of meat to meet their nutritional needs.

Cats are known for their agility, sharp senses, and predatory instincts. They have retractable claws, which they use for hunting and self-defense. Cats also have a keen sense of smell, hearing, and vision, which allow them to detect prey and navigate their environment.

In medical terms, cats can be hosts to various parasites and diseases that can affect humans and other animals. Some common feline diseases include rabies, feline leukemia virus (FeLV), feline immunodeficiency virus (FIV), and toxoplasmosis. It is important for cat owners to keep their pets healthy and up-to-date on vaccinations and preventative treatments to protect both the cats and their human companions.

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.

Cytomegalovirus (CMV) is a type of herpesvirus that can cause infection in humans. It is characterized by the enlargement of infected cells (cytomegaly) and is typically transmitted through close contact with an infected person, such as through saliva, urine, breast milk, or sexual contact.

CMV infection can also be acquired through organ transplantation, blood transfusions, or during pregnancy from mother to fetus. While many people infected with CMV experience no symptoms, it can cause serious complications in individuals with weakened immune systems, such as those undergoing cancer treatment or those who have HIV/AIDS.

In newborns, congenital CMV infection can lead to hearing loss, vision problems, and developmental delays. Pregnant women who become infected with CMV for the first time during pregnancy are at higher risk of transmitting the virus to their unborn child. There is no cure for CMV, but antiviral medications can help manage symptoms and reduce the risk of complications in severe cases.

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.

Hemocyanin is a copper-containing protein found in the blood of some mollusks and arthropods, responsible for oxygen transport. Unlike hemoglobin in vertebrates, which uses iron to bind oxygen, hemocyanins have copper ions that reversibly bind to oxygen, turning the blood blue when oxygenated. When deoxygenated, the color of the blood is pale blue-gray. Hemocyanins are typically found in a multi-subunit form and are released into the hemolymph (the equivalent of blood in vertebrates) upon exposure to air or oxygen. They play a crucial role in supplying oxygen to various tissues and organs within these invertebrate organisms.

Chronic lymphocytic leukemia (CLL) is a type of cancer that starts from cells that become certain white blood cells (called lymphocytes) in the bone marrow. The cancer (leukemia) cells start in the bone marrow but then go into the blood.

In CLL, the leukemia cells often build up slowly. Many people don't have any symptoms for at least a few years. But over time, the cells can spread to other parts of the body, including the lymph nodes, liver, and spleen.

The "B-cell" part of the name refers to the fact that the cancer starts in a type of white blood cell called a B lymphocyte or B cell. The "chronic" part means that this leukemia usually progresses more slowly than other types of leukemia.

It's important to note that chronic lymphocytic leukemia is different from chronic myelogenous leukemia (CML). Although both are cancers of the white blood cells, they start in different types of white blood cells and progress differently.

Vaccination is a simple, safe, and effective way to protect people against harmful diseases, before they come into contact with them. It uses your body's natural defenses to build protection to specific infections and makes your immune system stronger.

A vaccination usually contains a small, harmless piece of a virus or bacteria (or toxins produced by these germs) that has been made inactive or weakened so it won't cause the disease itself. This piece of the germ is known as an antigen. When the vaccine is introduced into the body, the immune system recognizes the antigen as foreign and produces antibodies to fight it.

If a person then comes into contact with the actual disease-causing germ, their immune system will recognize it and immediately produce antibodies to destroy it. The person is therefore protected against that disease. This is known as active immunity.

Vaccinations are important for both individual and public health. They prevent the spread of contagious diseases and protect vulnerable members of the population, such as young children, the elderly, and people with weakened immune systems who cannot be vaccinated or for whom vaccination is not effective.

Integrin α4β1, also known as Very Late Antigen-4 (VLA-4), is a heterodimeric transmembrane receptor protein composed of two subunits, α4 and β1. It is involved in various cellular activities such as adhesion, migration, and signaling. This integrin plays a crucial role in the immune system by mediating the interaction between leukocytes (white blood cells) and the endothelial cells that line blood vessels. The activation of Integrin α4β1 allows leukocytes to roll along and then firmly adhere to the endothelium, followed by their migration into surrounding tissues, particularly during inflammation and immune responses. Additionally, Integrin α4β1 also interacts with extracellular matrix proteins such as fibronectin and helps regulate cell survival, proliferation, and differentiation in various cell types.

Interphase is a phase in the cell cycle during which the cell primarily performs its functions of growth and DNA replication. It is the longest phase of the cell cycle, consisting of G1 phase (during which the cell grows and prepares for DNA replication), S phase (during which DNA replication occurs), and G2 phase (during which the cell grows further and prepares for mitosis). During interphase, the chromosomes are in their relaxed, extended form and are not visible under the microscope. Interphase is followed by mitosis, during which the chromosomes condense and separate to form two genetically identical daughter cells.

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.

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.

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.

Leukemia, T-cell is a type of cancer that affects the T-cells or T-lymphocytes, which are a type of white blood cells responsible for cell-mediated immunity. It is characterized by an excessive and uncontrolled production of abnormal T-cells in the bone marrow, leading to the displacement of healthy cells and impairing the body's ability to fight infections and regulate immune responses.

T-cell leukemia can be acute or chronic, depending on the rate at which the disease progresses. Acute T-cell leukemia progresses rapidly, while chronic T-cell leukemia has a slower course of progression. Symptoms may include fatigue, fever, frequent infections, weight loss, easy bruising or bleeding, and swollen lymph nodes. Treatment typically involves chemotherapy, radiation therapy, stem cell transplantation, or targeted therapy, depending on the type and stage of the disease.

A cohort study is a type of observational study in which a group of individuals who share a common characteristic or exposure are followed up over time to determine the incidence of a specific outcome or outcomes. The cohort, or group, is defined based on the exposure status (e.g., exposed vs. unexposed) and then monitored prospectively to assess for the development of new health events or conditions.

Cohort studies can be either prospective or retrospective in design. In a prospective cohort study, participants are enrolled and followed forward in time from the beginning of the study. In contrast, in a retrospective cohort study, researchers identify a cohort that has already been assembled through medical records, insurance claims, or other sources and then look back in time to assess exposure status and health outcomes.

Cohort studies are useful for establishing causality between an exposure and an outcome because they allow researchers to observe the temporal relationship between the two. They can also provide information on the incidence of a disease or condition in different populations, which can be used to inform public health policy and interventions. However, cohort studies can be expensive and time-consuming to conduct, and they may be subject to bias if participants are not representative of the population or if there is loss to follow-up.

HLA-B7 antigen is a type of human leukocyte antigen (HLA) found on the surface of cells in our body. The HLAs are proteins that help our immune system recognize and fight off foreign substances, such as viruses and bacteria. Specifically, HLA-B7 is a class I HLA antigen, which presents peptides from inside the cell to CD8+ T cells, a type of white blood cell that plays a crucial role in the immune response.

HLA-B7 has been identified as one of the many different HLA types that can be inherited from our parents. It is located on chromosome 6 and has several subtypes. The HLA-B7 antigen is associated with certain diseases, such as ankylosing spondylitis, a type of arthritis that affects the spine. However, having this HLA type does not necessarily mean that a person will develop the disease, as other genetic and environmental factors are also involved.

It's important to note that HLA typing is used in organ transplantation to match donors and recipients and reduce the risk of rejection. Knowing a patient's HLA type can help identify compatible donors and improve the chances of a successful transplant.

'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.

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.

Integrins are a family of cell-surface receptors that play crucial roles in various biological processes, including cell adhesion, migration, and signaling. Integrin alpha chains are one of the two subunits that make up an integrin heterodimer, with the other subunit being an integrin beta chain.

Integrin alpha chains are transmembrane glycoproteins consisting of a large extracellular domain, a single transmembrane segment, and a short cytoplasmic tail. The extracellular domain contains several domains that mediate ligand binding, while the cytoplasmic tail interacts with various cytoskeletal proteins and signaling molecules to regulate intracellular signaling pathways.

There are 18 different integrin alpha chains known in humans, each of which can pair with one or more beta chains to form distinct integrin heterodimers. These heterodimers exhibit unique ligand specificities and functions, allowing them to mediate diverse cell-matrix and cell-cell interactions.

In summary, integrin alpha chains are essential subunits of integrin receptors that play crucial roles in regulating cell adhesion, migration, and signaling by mediating interactions between cells and their extracellular environment.

A chimera, in the context of medicine and biology, is a single organism that is composed of cells with different genetics. This can occur naturally in some situations, such as when fraternal twins do not fully separate in utero and end up sharing some organs or tissues. The term "chimera" can also refer to an organism that contains cells from two different species, which can happen in certain types of genetic research or medical treatments. For example, a patient's cells might be genetically modified in a lab and then introduced into their body to treat a disease; if some of these modified cells mix with the patient's original cells, the result could be a chimera.

It's worth noting that the term "chimera" comes from Greek mythology, where it referred to a fire-breathing monster that was part lion, part goat, and part snake. In modern scientific usage, the term has a specific technical meaning related to genetics and organisms, but it may still evoke images of fantastical creatures for some people.

CD1 antigens are a group of molecules found on the surface of certain immune cells, including dendritic cells and B cells. They play a role in the immune system by presenting lipid antigens to T cells, which helps initiate an immune response against foreign substances such as bacteria and viruses. CD1 molecules are distinct from other antigen-presenting molecules like HLA because they present lipids rather than peptides. There are five different types of CD1 molecules (CD1a, CD1b, CD1c, CD1d, and CD1e) that differ in their tissue distribution and the types of lipid antigens they present.

Autologous transplantation is a medical procedure where cells, tissues, or organs are removed from a person, stored and then returned back to the same individual at a later time. This is different from allogeneic transplantation where the tissue or organ is obtained from another donor. The term "autologous" is derived from the Greek words "auto" meaning self and "logos" meaning study.

In autologous transplantation, the patient's own cells or tissues are used to replace or repair damaged or diseased ones. This reduces the risk of rejection and eliminates the need for immunosuppressive drugs, which are required in allogeneic transplants to prevent the body from attacking the foreign tissue.

Examples of autologous transplantation include:

* Autologous bone marrow or stem cell transplantation, where stem cells are removed from the patient's blood or bone marrow, stored and then reinfused back into the same individual after high-dose chemotherapy or radiation therapy to treat cancer.
* Autologous skin grafting, where a piece of skin is taken from one part of the body and transplanted to another area on the same person.
* Autologous chondrocyte implantation, where cartilage cells are harvested from the patient's own knee, cultured in a laboratory and then implanted back into the knee to repair damaged cartilage.

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.

Treatment outcome is a term used to describe the result or effect of medical treatment on a patient's health status. It can be measured in various ways, such as through symptoms improvement, disease remission, reduced disability, improved quality of life, or survival rates. The treatment outcome helps healthcare providers evaluate the effectiveness of a particular treatment plan and make informed decisions about future care. It is also used in clinical research to compare the efficacy of different treatments and improve patient care.

The cell cycle is a series of events that take place in a cell leading to its division and duplication. It consists of four main phases: G1 phase, S phase, G2 phase, and M phase.

During the G1 phase, the cell grows in size and synthesizes mRNA and proteins in preparation for DNA replication. In the S phase, the cell's DNA is copied, resulting in two complete sets of chromosomes. During the G2 phase, the cell continues to grow and produces more proteins and organelles necessary for cell division.

The M phase is the final stage of the cell cycle and consists of mitosis (nuclear division) and cytokinesis (cytoplasmic division). Mitosis results in two genetically identical daughter nuclei, while cytokinesis divides the cytoplasm and creates two separate daughter cells.

The cell cycle is regulated by various checkpoints that ensure the proper completion of each phase before progressing to the next. These checkpoints help prevent errors in DNA replication and division, which can lead to mutations and cancer.

Superantigens are a unique group of antigens that can cause widespread activation of the immune system. They are capable of stimulating large numbers of T-cells (a type of white blood cell) leading to massive cytokine release, which can result in a variety of symptoms such as fever, rash, and potentially life-threatening conditions like toxic shock syndrome. Superantigens are often produced by certain bacteria and viruses. They differ from traditional antigens because they do not need to be processed and presented by antigen-presenting cells to activate T-cells; instead, they directly bind to the major histocompatibility complex class II molecules and the T-cell receptor's variable region, leading to polyclonal T-cell activation.

Infectious Mononucleosis, also known as "mono" or the "kissing disease," is a common infectious illness caused by the Epstein-Barr virus (EBV). It primarily affects adolescents and young adults. The medical definition of Infectious Mononucleosis includes the following signs and symptoms:

1. Infection: Infectious Mononucleosis is an infection that spreads through saliva, hence the nickname "kissing disease." It can also be transmitted through sharing food, drinks, or personal items such as toothbrushes or utensils with an infected person.
2. Incubation period: The incubation period for Infectious Mononucleosis is typically 4-6 weeks after exposure to the virus.
3. Symptoms: Common symptoms of Infectious Mononucleosis include fever, sore throat (often severe and may resemble strep throat), fatigue, swollen lymph nodes (particularly in the neck and armpits), and skin rash (in some cases).
4. Diagnosis: The diagnosis of Infectious Mononucleosis is typically made based on a combination of clinical symptoms, physical examination findings, and laboratory test results. A complete blood count (CBC) may reveal an increased number of white blood cells, particularly atypical lymphocytes. Additionally, the Paul-Bunnell or Monospot test can detect heterophile antibodies, which are present in about 85% of cases after the first week of illness.
5. Treatment: There is no specific antiviral treatment for Infectious Mononucleosis. Management typically involves supportive care, such as rest, hydration, and pain relief for symptoms like sore throat and fever.
6. Complications: Although most cases of Infectious Mononucleosis resolve without significant complications, some individuals may experience complications such as splenomegaly (enlarged spleen), hepatitis, or neurological issues. Rarely, the virus can cause more severe complications like myocarditis (inflammation of the heart muscle) or hemolytic anemia (destruction of red blood cells).
7. Prevention: Preventing Infectious Mononucleosis is difficult since it is primarily spread through respiratory droplets and saliva. However, practicing good hygiene, such as covering the mouth and nose when coughing or sneezing and avoiding sharing personal items like utensils or drinking glasses, can help reduce the risk of transmission.

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.

Beta-2 microglobulin (β2M) is a small protein that is a component of the major histocompatibility complex class I molecule, which plays a crucial role in the immune system. It is found on the surface of almost all nucleated cells in the body and is involved in presenting intracellular peptides to T-cells for immune surveillance.

β2M is produced at a relatively constant rate by cells throughout the body and is freely filtered by the glomeruli in the kidneys. Under normal circumstances, most of the filtrated β2M is reabsorbed and catabolized in the proximal tubules of the nephrons. However, when the glomerular filtration rate (GFR) is decreased, as in chronic kidney disease (CKD), the reabsorption capacity of the proximal tubules becomes overwhelmed, leading to increased levels of β2M in the blood and its subsequent appearance in the urine.

Elevated serum and urinary β2M levels have been associated with various clinical conditions, such as CKD, multiple myeloma, autoimmune disorders, and certain infectious diseases. Measuring β2M concentrations can provide valuable information for diagnostic, prognostic, and monitoring purposes in these contexts.

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.

Kidney transplantation is a surgical procedure where a healthy kidney from a deceased or living donor is implanted into a patient with end-stage renal disease (ESRD) or permanent kidney failure. The new kidney takes over the functions of filtering waste and excess fluids from the blood, producing urine, and maintaining the body's electrolyte balance.

The transplanted kidney is typically placed in the lower abdomen, with its blood vessels connected to the recipient's iliac artery and vein. The ureter of the new kidney is then attached to the recipient's bladder to ensure proper urine flow. Following the surgery, the patient will require lifelong immunosuppressive therapy to prevent rejection of the transplanted organ by their immune system.

Cyclosporine is a medication that belongs to a class of drugs called immunosuppressants. It is primarily used to prevent the rejection of transplanted organs, such as kidneys, livers, and hearts. Cyclosporine works by suppressing the activity of the immune system, which helps to reduce the risk of the body attacking the transplanted organ.

In addition to its use in organ transplantation, cyclosporine may also be used to treat certain autoimmune diseases, such as rheumatoid arthritis and psoriasis. It does this by suppressing the overactive immune response that contributes to these conditions.

Cyclosporine is available in capsule, oral solution, and injectable forms. Common side effects of the medication include kidney problems, high blood pressure, tremors, headache, and nausea. Long-term use of cyclosporine can also increase the risk of certain types of cancer and infections.

It is important to note that cyclosporine should only be used under the close supervision of a healthcare provider, as it requires regular monitoring of blood levels and kidney function.

HIV antigens refer to the proteins present on the surface or within the human immunodeficiency virus (HIV), which can stimulate an immune response in the infected individual. These antigens are recognized by the host's immune system, specifically by CD4+ T cells and antibodies, leading to their activation and production. Two significant HIV antigens are the HIV-1 p24 antigen and the gp120/gp41 envelope proteins. The p24 antigen is a capsid protein found within the viral particle, while the gp120/gp41 complex forms the viral envelope and facilitates viral entry into host cells. Detection of HIV antigens in clinical settings, such as in the ELISA or Western blot tests, helps diagnose HIV infection and monitor disease progression.

Retroviridae is a family of viruses that includes human immunodeficiency virus (HIV) and other viruses that primarily use RNA as their genetic material. The name "retrovirus" comes from the fact that these viruses reverse transcribe their RNA genome into DNA, which then becomes integrated into the host cell's genome. This is a unique characteristic of retroviruses, as most other viruses use DNA as their genetic material.

Retroviruses can cause a variety of diseases in animals and humans, including cancer, neurological disorders, and immunodeficiency syndromes like AIDS. They have a lipid membrane envelope that contains glycoprotein spikes, which allow them to attach to and enter host cells. Once inside the host cell, the viral RNA is reverse transcribed into DNA by the enzyme reverse transcriptase, which is then integrated into the host genome by the enzyme integrase.

Retroviruses can remain dormant in the host genome for extended periods of time, and may be reactivated under certain conditions to produce new viral particles. This ability to integrate into the host genome has also made retroviruses useful tools in molecular biology, where they are used as vectors for gene therapy and other genetic manipulations.

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.

"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.

Thymoma is a type of tumor that originates from the thymus gland, which is a part of the immune system located in the chest behind the breastbone. Thymomas are typically slow-growing and often do not cause any symptoms until they have grown quite large or spread to other parts of the body.

Thymomas can be classified into different types based on their appearance under a microscope, such as type A, AB, B1, B2, and B3. These classifications are important because they can help predict how aggressive the tumor is likely to be and how it should be treated.

Symptoms of thymoma may include cough, chest pain, difficulty breathing, or swelling in the face or neck. Thymomas can also be associated with autoimmune disorders such as myasthenia gravis, which affects muscle strength and mobility. Treatment for thymoma typically involves surgical removal of the tumor, often followed by radiation therapy or chemotherapy to help prevent recurrence.

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.

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.

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.

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.

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.

Catecholamines are a group of hormones and neurotransmitters that are derived from the amino acid tyrosine. The most well-known catecholamines are dopamine, norepinephrine (also known as noradrenaline), and epinephrine (also known as adrenaline). These hormones are produced by the adrenal glands and are released into the bloodstream in response to stress. They play important roles in the "fight or flight" response, increasing heart rate, blood pressure, and alertness. In addition to their role as hormones, catecholamines also function as neurotransmitters, transmitting signals in the nervous system. Disorders of catecholamine regulation can lead to a variety of medical conditions, including hypertension, mood disorders, and neurological disorders.

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.

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.

Herpesvirus 2, Saimiriine (SaHV-2) is a species of herpesvirus that primarily infects the primate species Saimiri sciureus, also known as the squirrel monkey. It is a member of the genus Rhadinovirus in the subfamily Gammaherpesvirinae. SaHV-2 has been associated with lymphoproliferative diseases and lymphomas in its natural host. The virus has a complex structure, consisting of an outer envelope, a protein layer called the capsid, and a DNA genome. It employs a sophisticated replication strategy to establish latency and evade the host's immune response.

It is important to note that SaHV-2 does not infect humans and is primarily studied in the context of comparative primatology and viral pathogenesis research.

A micronucleus test is a type of genetic toxicology assay used to detect the presence of micronuclei in cells, which are small chromosomal fragments or whole chromosomes that have been missegregated during cell division. The test measures the frequency of micronuclei in cells exposed to a potential genotoxic agent, such as a chemical or radiation, and compares it to the frequency in untreated control cells.

The assay is typically performed on cultured mammalian cells, such as human lymphocytes or Chinese hamster ovary (CHO) cells, and involves exposing the cells to the test agent for a specific period of time, followed by staining and examination of the cells under a microscope. The micronuclei are identified based on their size, shape, and staining characteristics, and the frequency of micronucleated cells is calculated as a measure of genotoxic potential.

Micronucleus tests are widely used in regulatory toxicology to assess the genetic safety of chemicals, drugs, and other substances, and can provide valuable information on potential risks to human health. The test is also used in basic research to study the mechanisms of genotoxicity and chromosomal instability.

Viral proteins are the proteins that are encoded by the viral genome and are essential for the viral life cycle. These proteins can be structural or non-structural and play various roles in the virus's replication, infection, and assembly process. Structural proteins make up the physical structure of the virus, including the capsid (the protein shell that surrounds the viral genome) and any envelope proteins (that may be present on enveloped viruses). Non-structural proteins are involved in the replication of the viral genome and modulation of the host cell environment to favor viral replication. Overall, a thorough understanding of viral proteins is crucial for developing antiviral therapies and vaccines.

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.

Immunoglobulin D (IgD) is a type of antibody that is present in the blood and other bodily fluids. It is one of the five classes of immunoglobulins (IgA, IgD, IgE, IgG, and IgM) found in humans and plays a role in the immune response.

IgD is produced by B cells, a type of white blood cell that is responsible for producing antibodies. It is primarily found on the surface of mature B cells, where it functions as a receptor for antigens (foreign substances that trigger an immune response). When an antigen binds to IgD on the surface of a B cell, it activates the B cell and stimulates it to produce and secrete antibodies specific to that antigen.

IgD is found in relatively low concentrations in the blood compared to other immunoglobulins, and its precise functions are not fully understood. However, it is thought to play a role in the regulation of B cell activation and the immune response. Additionally, some research suggests that IgD may have a direct role in protecting against certain types of infections.

It's worth noting that genetic deficiencies in IgD are not typically associated with any significant immunological abnormalities or increased susceptibility to infection.

Nuclear proteins are a category of proteins that are primarily found in the nucleus of a eukaryotic cell. They play crucial roles in various nuclear functions, such as DNA replication, transcription, repair, and RNA processing. This group includes structural proteins like lamins, which form the nuclear lamina, and regulatory proteins, such as histones and transcription factors, that are involved in gene expression. Nuclear localization signals (NLS) often help target these proteins to the nucleus by interacting with importin proteins during active transport across the nuclear membrane.

Synthetic vaccines are artificially produced, designed to stimulate an immune response and provide protection against specific diseases. Unlike traditional vaccines that are derived from weakened or killed pathogens, synthetic vaccines are created using synthetic components, such as synthesized viral proteins, DNA, or RNA. These components mimic the disease-causing agent and trigger an immune response without causing the actual disease. The use of synthetic vaccines offers advantages in terms of safety, consistency, and scalability in production, making them valuable tools for preventing infectious diseases.

Clonal anergy is a term used in immunology to describe a state of immune tolerance or unresponsiveness in certain T cells, a type of white blood cell that plays a central role in the body's immune response. This condition arises when T cells are exposed to persistent antigens, such as those derived from viruses or tumors, and fail to become fully activated.

In normal circumstances, when a T cell encounters an antigen presented by an antigen-presenting cell (APC), it becomes activated and undergoes clonal expansion, producing many copies of itself that are specific for that particular antigen. These activated T cells then migrate to the site of infection or tissue damage and help coordinate the immune response to eliminate the threat.

However, in some cases, persistent exposure to an antigen can lead to a state of exhaustion or anergy in the T cells, where they are no longer able to respond effectively to that antigen. This is thought to occur due to chronic stimulation and activation of the T cells, which can lead to the upregulation of inhibitory receptors and the downregulation of activating receptors on their surface.

Clonal anergy is a mechanism by which the immune system attempts to prevent excessive or inappropriate immune responses that could cause tissue damage or autoimmunity. However, it can also be a barrier to effective immunotherapy for diseases such as cancer, where T cells need to be activated and able to recognize and eliminate tumor cells.

In summary, clonal anergy is a state of immune tolerance in certain T cells that have been persistently exposed to antigens, leading to their failure to become fully activated and respond effectively to those antigens.

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.

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.

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.

Influenza A virus is defined as a negative-sense, single-stranded, segmented RNA virus belonging to the family Orthomyxoviridae. It is responsible for causing epidemic and pandemic influenza in humans and is also known to infect various animal species, such as birds, pigs, horses, and seals. The viral surface proteins, hemagglutinin (HA) and neuraminidase (NA), are the primary targets for antiviral drugs and vaccines. There are 18 different HA subtypes and 11 known NA subtypes, which contribute to the diversity and antigenic drift of Influenza A viruses. The zoonotic nature of this virus allows for genetic reassortment between human and animal strains, leading to the emergence of novel variants with pandemic potential.

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.

Chemokine (C-C motif) ligand 19 (CCL19), also known as macrophage inflammatory protein-3 beta (MIP-3β) or exodus-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 immunity and inflammation by directing the migration of various immune cells to sites of infection, injury, or inflammation through a process called chemotaxis.

CCL19 is primarily produced by mature dendritic cells, a type of antigen-presenting cell that plays a key role in initiating and regulating adaptive immunity. CCL19 attracts various immune cells expressing its receptor, CCR7, including T cells, B cells, and dendritic cells, to the T cell zones of secondary lymphoid organs such as lymph nodes and spleen. This facilitates the encounter between antigen-presenting cells and T cells, leading to the activation of T cells and the generation of adaptive immune responses.

In addition to its role in immunity and inflammation, CCL19 has been implicated in various physiological and pathological processes, such as lymphoid organ development, angiogenesis, and cancer metastasis. Dysregulation of CCL19 expression or function has been associated with several diseases, including autoimmune disorders, chronic inflammation, and malignancies.

Viral DNA refers to the genetic material present in viruses that consist of DNA as their core component. Deoxyribonucleic acid (DNA) is one of the two types of nucleic acids that are responsible for storing and transmitting genetic information in living organisms. Viruses are infectious agents much smaller than bacteria that can only replicate inside the cells of other organisms, called hosts.

Viral DNA can be double-stranded (dsDNA) or single-stranded (ssDNA), depending on the type of virus. Double-stranded DNA viruses have a genome made up of two complementary strands of DNA, while single-stranded DNA viruses contain only one strand of DNA.

Examples of dsDNA viruses include Adenoviruses, Herpesviruses, and Poxviruses, while ssDNA viruses include Parvoviruses and Circoviruses. Viral DNA plays a crucial role in the replication cycle of the virus, encoding for various proteins necessary for its multiplication and survival within the host cell.

HLA-B antigens are human leukocyte antigen (HLA) proteins found on the surface of cells that play an important role in the body's immune system. They are part of the major histocompatibility complex (MHC) class I molecules, which present pieces of proteins from inside the cell to T-cells, a type of white blood cell involved in immune responses.

HLA-B antigens are highly polymorphic, meaning that there are many different variations or alleles of this gene in the human population. This genetic diversity allows for a wide range of potential HLA-B proteins to be expressed, which can help recognize and respond to a variety of foreign substances, such as viruses and cancer cells.

The HLA-B antigens are inherited from both parents, and an individual may express one or two different HLA-B antigens depending on their genetic makeup. The specific combination of HLA-B antigens that a person expresses can have implications for their susceptibility to certain diseases, as well as their compatibility with organ transplants.

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.

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.

T-cell receptor beta (TCRβ) is a type of protein found on the surface of certain immune cells called T cells. These receptors play a critical role in the adaptive immune response, enabling T cells to recognize and respond to specific antigens presented by other cells in the body. The TCRβ chain is one of the two polypeptide chains that make up the T-cell receptor complex, with the other being the TCR alpha (TCRα) chain.

Genes related to TCRβ are located within a region of the human genome known as the T-cell receptor beta locus, which spans approximately 600 kilobases on chromosome 7 (7q34). This locus contains around 58 variable (V), 2 diversity (D), and 13 joining (J) gene segments, along with a constant (C) region. During the development of T cells in the thymus, a process called V(D)J recombination occurs, where one V, one D, and one J segment are randomly selected and joined together to form a unique variable region exon that encodes the antigen-binding site of the TCRβ protein. This diversification mechanism allows for the recognition of a vast array of different antigens, contributing to the specificity and adaptability of the immune response.

Cell aggregation is the process by which individual cells come together and adhere to each other to form a group or cluster. This phenomenon can occur naturally during embryonic development, tissue repair, and wound healing, as well as in the formation of multicellular organisms such as slime molds. In some cases, cell aggregation may also be induced in the laboratory setting through the use of various techniques, including the use of cell culture surfaces that promote cell-to-cell adhesion or the addition of factors that stimulate the expression of adhesion molecules on the cell surface.

Cell aggregation can be influenced by a variety of factors, including the type and properties of the cells involved, as well as environmental conditions such as pH, temperature, and nutrient availability. The ability of cells to aggregate is often mediated by the presence of adhesion molecules on the cell surface, such as cadherins, integrins, and immunoglobulin-like cell adhesion molecules (Ig-CAMs). These molecules interact with each other and with extracellular matrix components to promote cell-to-cell adhesion and maintain the stability of the aggregate.

In some contexts, abnormal or excessive cell aggregation can contribute to the development of diseases such as cancer, fibrosis, and inflammatory disorders. For example, the aggregation of cancer cells can facilitate their invasion and metastasis, while the accumulation of fibrotic cells in tissues can lead to organ dysfunction and failure. Understanding the mechanisms that regulate cell aggregation is therefore an important area of research with potential implications for the development of new therapies and treatments for a variety of 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.

Histocompatibility testing, also known as tissue typing, is a medical procedure that determines the compatibility of tissues between two individuals, usually a potential donor and a recipient for organ or bone marrow transplantation. The test identifies specific antigens, called human leukocyte antigens (HLAs), found on the surface of most cells in the body. These antigens help the immune system distinguish between "self" and "non-self" cells.

The goal of histocompatibility testing is to find a donor whose HLA markers closely match those of the recipient, reducing the risk of rejection of the transplanted organ or tissue. The test involves taking blood samples from both the donor and the recipient and analyzing them for the presence of specific HLA antigens using various laboratory techniques such as molecular typing or serological testing.

A high degree of histocompatibility between the donor and recipient is crucial to ensure the success of the transplantation procedure, minimize complications, and improve long-term outcomes.

Proto-oncogene proteins c-bcl-2 are a group of proteins that play a role in regulating cell death (apoptosis). The c-bcl-2 gene produces one of these proteins, which helps to prevent cells from undergoing apoptosis. This protein is located on the membrane of mitochondria and endoplasmic reticulum and it can inhibit the release of cytochrome c, a key player in the activation of caspases, which are enzymes that trigger apoptosis.

In normal cells, the regulation of c-bcl-2 protein helps to maintain a balance between cell proliferation and cell death, ensuring proper tissue homeostasis. However, when the c-bcl-2 gene is mutated or its expression is dysregulated, it can contribute to cancer development by allowing cancer cells to survive and proliferate. High levels of c-bcl-2 protein have been found in many types of cancer, including leukemia, lymphoma, and carcinomas, and are often associated with a poor prognosis.

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.

Suppressor factors, immunologic, refer to substances that can suppress or decrease the immune response. They were first described in the 1970s and are produced by certain cells of the immune system, such as T cells. Suppressor factors help to maintain immune homeostasis and prevent overactive immune responses that can lead to autoimmune diseases or chronic inflammation.

Immunologic suppressor factors can inhibit the activation and proliferation of various immune cells, including T cells, B cells, and natural killer (NK) cells. They can also suppress the production of cytokines, which are signaling molecules that help regulate the immune response. Suppressor factors have been studied in the context of various diseases, including cancer, autoimmune disorders, and transplant rejection.

However, the concept of immunologic suppressor factors has been controversial, and their precise mechanisms of action are not fully understood. Some researchers have questioned whether they truly exist as distinct entities or whether they represent a heterogeneous group of regulatory molecules with diverse functions. Nonetheless, the study of immunologic suppressor factors remains an active area of research, as understanding how they work could lead to new therapies for a variety of diseases.

The nef gene in the Human Immunodeficiency Virus (HIV) encodes for the nef protein, which is a key regulatory protein for the virus. The nef gene products, which include the nef protein and its cleavage fragments, play several crucial roles in the viral life cycle and the pathogenesis of HIV infection.

The nef protein is a myristoylated, multifunctional type I transmembrane protein that localizes to the plasma membrane and endosomal compartments. It has been shown to have several effects on both viral replication and host cell functions:

1. Downregulation of CD4 receptor and major histocompatibility complex class I (MHC-I) molecules from the cell surface: By reducing the expression of these molecules, nef helps HIV to evade the immune response and enhances viral infectivity.
2. Enhancement of virion infectivity: Nef can increase the incorporation of viral envelope proteins into virions and promote their fusogenic activity, leading to more efficient infection of target cells.
3. Augmentation of viral replication: Nef contributes to the activation of signaling pathways that stimulate viral gene expression and support the establishment of viral reservoirs in infected cells.
4. Modulation of host cell signal transduction: Nef can interact with various host cell proteins, affecting their functions and contributing to HIV-induced immune dysfunction and disease progression.

The nef gene products are essential for efficient HIV replication and pathogenesis, making them potential targets for antiretroviral therapy and vaccine development.

Interleukin-16 (IL-16) is a chemokine, which is a type of signaling protein involved in immune responses and inflammation. IL-16 was initially identified as a T cell chemoattractant, meaning it can attract or draw T cells, a type of white blood cell, to areas where it is produced.

IL-16 is produced by a variety of cells, including CD4+ T cells, eosinophils, mast cells, and epithelial cells. It is involved in the regulation of immune responses, including the activation and proliferation of T cells, as well as the recruitment of other immune cells to sites of inflammation or injury.

IL-16 binds to a specific receptor called CD4, which is found on the surface of certain immune cells, including T cells, monocytes, and dendritic cells. The binding of IL-16 to its receptor triggers a series of intracellular signaling events that ultimately lead to changes in gene expression and cell behavior.

In addition to its role in the immune system, IL-16 has also been implicated in various disease processes, including asthma, allergies, autoimmune disorders, and cancer.

Tetanus toxoid is a purified and inactivated form of the tetanus toxin, which is derived from the bacterium Clostridium tetani. It is used as a vaccine to induce active immunity against tetanus, a potentially fatal disease caused by this toxin. The toxoid is produced through a series of chemical treatments that modify the toxic properties of the tetanus toxin while preserving its antigenic qualities. This allows the immune system to recognize and develop protective antibodies against the toxin without causing illness. Tetanus toxoid is often combined with diphtheria and/or pertussis toxoids in vaccines such as DTaP, Tdap, and Td.

Inbred NOD (Nonobese Diabetic) mice are a strain of laboratory mice that are genetically predisposed to develop autoimmune diabetes. This strain was originally developed in Japan and has been widely used as an animal model for studying type 1 diabetes and its complications.

NOD mice typically develop diabetes spontaneously at around 12-14 weeks of age, although the onset and severity of the disease can vary between individual mice. The disease is caused by a breakdown in immune tolerance, leading to an autoimmune attack on the insulin-producing beta cells of the pancreas.

Inbred NOD mice are highly valuable for research purposes because they exhibit many of the same genetic and immunological features as human patients with type 1 diabetes. By studying these mice, researchers can gain insights into the underlying mechanisms of the disease and develop new treatments and therapies.

Synovial fluid is a viscous, clear, and straw-colored fluid found in the cavities of synovial joints, bursae, and tendon sheaths. It is produced by the synovial membrane, which lines the inner surface of the capsule surrounding these structures.

The primary function of synovial fluid is to reduce friction between articulating surfaces, providing lubrication for smooth and painless movement. It also acts as a shock absorber, protecting the joints from external forces during physical activities. Synovial fluid contains nutrients that nourish the articular cartilage, hyaluronic acid, which provides its viscoelastic properties, and lubricin, a protein responsible for boundary lubrication.

Abnormalities in synovial fluid composition or volume can indicate joint-related disorders, such as osteoarthritis, rheumatoid arthritis, gout, infection, or trauma. Analysis of synovial fluid is often used diagnostically to determine the underlying cause of joint pain, inflammation, or dysfunction.

Histocompatibility antigen H-2D is a type of major histocompatibility complex (MHC) class I molecule found in mice. It is a transmembrane protein located on the surface of nucleated cells, which plays a crucial role in the adaptive immune system. The primary function of H-2D is to present endogenous peptide antigens to CD8+ T cells, also known as cytotoxic T lymphocytes (CTLs).

H-2D molecules are encoded by genes within the H-2D region of the MHC on chromosome 17. These genes have multiple alleles, resulting in a high degree of polymorphism, which contributes to the diversity of the immune response among different mouse strains. The peptide-binding groove of H-2D molecules is formed by two alpha helices and eight beta pleats, creating a specific binding site for antigenic peptides.

The peptides presented by H-2D molecules are derived from intracellular proteins that undergo degradation in the proteasome. These peptides are then transported into the endoplasmic reticulum, where they bind to H-2D molecules with the assistance of chaperone proteins like tapasin and calreticulin. The H-2D-peptide complex is then transported to the cell surface for presentation to CD8+ T cells.

Recognition of H-2D-peptide complexes by CD8+ T cells leads to their activation, proliferation, and differentiation into effector CTLs. Activated CTLs can recognize and eliminate virus-infected or malignant cells displaying specific H-2D-peptide complexes, thereby playing a critical role in the cell-mediated immune response.

In summary, histocompatibility antigen H-2D is a polymorphic MHC class I molecule in mice that presents endogenous peptide antigens to CD8+ T cells, contributing significantly to the adaptive immune response and the elimination of infected or malignant cells.

Hodgkin disease, also known as Hodgkin lymphoma, is a type of cancer that originates in the white blood cells called lymphocytes. It typically affects the lymphatic system, which is a network of vessels and glands spread throughout the body. The disease is characterized by the presence of a specific type of abnormal cell, known as a Reed-Sternberg cell, within the affected lymph nodes.

The symptoms of Hodgkin disease may include painless swelling of the lymph nodes in the neck, armpits, or groin; fever; night sweats; weight loss; and fatigue. The exact cause of Hodgkin disease is unknown, but it is thought to involve a combination of genetic, environmental, and infectious factors.

Hodgkin disease is typically treated with a combination of chemotherapy, radiation therapy, and/or immunotherapy, depending on the stage and extent of the disease. With appropriate treatment, the prognosis for Hodgkin disease is generally very good, with a high cure rate. However, long-term side effects of treatment may include an increased risk of secondary cancers and other health problems.

Survival analysis is a branch of statistics that deals with the analysis of time to event data. It is used to estimate the time it takes for a certain event of interest to occur, such as death, disease recurrence, or treatment failure. The event of interest is called the "failure" event, and survival analysis estimates the probability of not experiencing the failure event until a certain point in time, also known as the "survival" probability.

Survival analysis can provide important information about the effectiveness of treatments, the prognosis of patients, and the identification of risk factors associated with the event of interest. It can handle censored data, which is common in medical research where some participants may drop out or be lost to follow-up before the event of interest occurs.

Survival analysis typically involves estimating the survival function, which describes the probability of surviving beyond a certain time point, as well as hazard functions, which describe the instantaneous rate of failure at a given time point. Other important concepts in survival analysis include median survival times, restricted mean survival times, and various statistical tests to compare survival curves between groups.

Experimental leukemia refers to the stage of research or clinical trials where new therapies, treatments, or diagnostic methods are being studied for leukemia. Leukemia is a type of cancer that affects the blood and bone marrow, leading to an overproduction of abnormal white blood cells.

In the experimental stage, researchers investigate various aspects of leukemia, such as its causes, progression, and potential treatments. They may conduct laboratory studies using cell cultures or animal models to understand the disease better and test new therapeutic approaches. Additionally, clinical trials may be conducted to evaluate the safety and efficacy of novel treatments in human patients with leukemia.

Experimental research in leukemia is crucial for advancing our understanding of the disease and developing more effective treatment strategies. It involves a rigorous and systematic process that adheres to ethical guidelines and scientific standards to ensure the validity and reliability of the findings.

K562 cells are a type of human cancer cell that are commonly used in scientific research. They are derived from a patient with chronic myelogenous leukemia (CML), a type of cancer that affects the blood and bone marrow.

K562 cells are often used as a model system to study various biological processes, including cell signaling, gene expression, differentiation, and apoptosis (programmed cell death). They are also commonly used in drug discovery and development, as they can be used to test the effectiveness of potential new therapies against cancer.

K562 cells have several characteristics that make them useful for research purposes. They are easy to grow and maintain in culture, and they can be manipulated genetically to express or knock down specific genes. Additionally, K562 cells are capable of differentiating into various cell types, such as red blood cells and megakaryocytes, which allows researchers to study the mechanisms of cell differentiation.

It's important to note that while K562 cells are a valuable tool for research, they do not fully recapitulate the complexity of human CML or other cancers. Therefore, findings from studies using K562 cells should be validated in more complex model systems or in clinical trials before they can be translated into treatments for patients.

Chemokine (C-X-C motif) ligand 12 (CXCL12), also known as stromal cell-derived factor 1 (SDF-1), is a small signaling protein belonging to the chemokine family. Chemokines are a group of cytokines, or signaling molecules, that play important roles in immune responses and inflammation by recruiting and activating various immune cells.

CXCL12 is produced by several types of cells, including stromal cells, endothelial cells, and certain immune cells. It exerts its effects by binding to a specific receptor called C-X-C chemokine receptor type 4 (CXCR4), which is found on the surface of various cell types, including immune cells, stem cells, and some cancer cells.

The CXCL12-CXCR4 axis plays crucial roles in various physiological processes, such as embryonic development, tissue homeostasis, hematopoiesis (the formation of blood cells), and neurogenesis (the formation of neurons). Additionally, this signaling pathway has been implicated in several pathological conditions, including cancer metastasis, inflammatory diseases, and HIV infection.

In summary, Chemokine CXCL12 is a small signaling protein that binds to the CXCR4 receptor and plays essential roles in various physiological processes and pathological conditions.

Follow-up studies are a type of longitudinal research that involve repeated observations or measurements of the same variables over a period of time, in order to understand their long-term effects or outcomes. In medical context, follow-up studies are often used to evaluate the safety and efficacy of medical treatments, interventions, or procedures.

In a typical follow-up study, a group of individuals (called a cohort) who have received a particular treatment or intervention are identified and then followed over time through periodic assessments or data collection. The data collected may include information on clinical outcomes, adverse events, changes in symptoms or functional status, and other relevant measures.

The results of follow-up studies can provide important insights into the long-term benefits and risks of medical interventions, as well as help to identify factors that may influence treatment effectiveness or patient outcomes. However, it is important to note that follow-up studies can be subject to various biases and limitations, such as loss to follow-up, recall bias, and changes in clinical practice over time, which must be carefully considered when interpreting the results.

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.

Skin transplantation, also known as skin grafting, is a surgical procedure that involves the removal of healthy skin from one part of the body (donor site) and its transfer to another site (recipient site) that has been damaged or lost due to various reasons such as burns, injuries, infections, or diseases. The transplanted skin can help in healing wounds, restoring functionality, and improving the cosmetic appearance of the affected area. There are different types of skin grafts, including split-thickness grafts, full-thickness grafts, and composite grafts, which vary in the depth and size of the skin removed and transplanted. The success of skin transplantation depends on various factors, including the size and location of the wound, the patient's overall health, and the availability of suitable donor sites.

Viral matrix proteins are structural proteins that play a crucial role in the morphogenesis and life cycle of many viruses. They are often located between the viral envelope and the viral genome, serving as a scaffold for virus assembly and budding. These proteins also interact with other viral components, such as the viral genome, capsid proteins, and envelope proteins, to form an infectious virion. Additionally, matrix proteins can have regulatory functions, influencing viral transcription, replication, and host cell responses. The specific functions of viral matrix proteins vary among different virus families.

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.

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.

CD44 is a type of protein found on the surface of some cells in the human body. It is a cell adhesion molecule and is involved in various biological processes such as cell-cell interaction, lymphocyte activation, and migration of cells. CD44 also acts as a receptor for hyaluronic acid, a component of the extracellular matrix.

As an antigen, CD44 can be recognized by certain immune cells, including T cells and B cells, and can play a role in the immune response. There are several isoforms of CD44 that exist due to alternative splicing of its mRNA, leading to differences in its structure and function.

CD44 has been studied in the context of cancer, where it can contribute to tumor growth, progression, and metastasis. In some cases, high levels of CD44 have been associated with poor prognosis in certain types of cancer. However, CD44 also has potential roles in tumor suppression and immune surveillance, making its overall role in cancer complex and context-dependent.

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.

'NK Cell Lectin-Like Receptor Subfamily B' refers to a group of genes that encode proteins found on natural killer (NK) cells, which are a type of white blood cell in the human body. These proteins belong to a larger family called C-type lectin receptors (CLRs), which are involved in various immune functions such as pathogen recognition and immune cell activation.

The NK Cell Lectin-Like Receptor Subfamily B includes several genes, such as NKp80, NKp46, and NKp30, that encode proteins expressed on the surface of NK cells. These proteins function as activating receptors, meaning they can trigger NK cell activation and subsequent immune responses when they bind to specific ligands on the surface of infected or abnormal cells.

Overall, the NK Cell Lectin-Like Receptor Subfamily B plays an essential role in the innate immune response against viral infections and cancer by mediating NK cell cytotoxicity and cytokine production.

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.

Immunoglobulin heavy chains are proteins that make up the framework of antibodies, which are Y-shaped immune proteins. These heavy chains, along with light chains, form the antigen-binding sites of an antibody, which recognize and bind to specific foreign substances (antigens) in order to neutralize or remove them from the body.

The heavy chain is composed of a variable region, which contains the antigen-binding site, and constant regions that determine the class and function of the antibody. There are five classes of immunoglobulins (IgA, IgD, IgE, IgG, and IgM) that differ in their heavy chain constant regions and therefore have different functions in the immune response.

Immunoglobulin heavy chains are synthesized by B cells, a type of white blood cell involved in the adaptive immune response. The genetic rearrangement of immunoglobulin heavy chain genes during B cell development results in the production of a vast array of different antibodies with unique antigen-binding sites, allowing for the recognition and elimination of a wide variety of pathogens.

Transplantation Immunology is a branch of medicine that deals with the immune responses occurring between a transplanted organ or tissue and the recipient's body. It involves understanding and managing the immune system's reaction to foreign tissue, which can lead to rejection of the transplanted organ. This field also studies the use of immunosuppressive drugs to prevent rejection and the potential risks and side effects associated with their use. The main goal of transplantation immunology is to find ways to promote the acceptance of transplanted tissue while minimizing the risk of infection and other complications.

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.

A viral vaccine is a biological preparation that introduces your body to a specific virus in a way that helps your immune system build up protection against the virus without causing the illness. Viral vaccines can be made from weakened or inactivated forms of the virus, or parts of the virus such as proteins or sugars. Once introduced to the body, the immune system recognizes the virus as foreign and produces an immune response, including the production of antibodies. These antibodies remain in the body and provide immunity against future infection with that specific virus.

Viral vaccines are important tools for preventing infectious diseases caused by viruses, such as influenza, measles, mumps, rubella, polio, hepatitis A and B, rabies, rotavirus, chickenpox, shingles, and some types of cancer. Vaccination programs have led to the control or elimination of many infectious diseases that were once common.

It's important to note that viral vaccines are not effective against bacterial infections, and separate vaccines must be developed for each type of virus. Additionally, because viruses can mutate over time, it is necessary to update some viral vaccines periodically to ensure continued protection.

Interleukin-5 (IL-5) is a type of cytokine, which is a small signaling protein that mediates and regulates immunity, inflammation, and hematopoiesis. IL-5 is primarily produced by activated T cells, especially Th2 cells, as well as mast cells, eosinophils, and innate lymphoid cells (ILCs).

The primary function of IL-5 is to regulate the growth, differentiation, activation, and survival of eosinophils, a type of white blood cell that plays a crucial role in the immune response against parasitic infections. IL-5 also enhances the ability of eosinophils to migrate from the bone marrow into the bloodstream and then into tissues, where they can participate in immune responses.

In addition to its effects on eosinophils, IL-5 has been shown to have a role in the regulation of B cell function, including promoting the survival and differentiation of B cells into antibody-secreting plasma cells. Dysregulation of IL-5 production and activity has been implicated in several diseases, including asthma, allergies, and certain parasitic infections.

Melanoma-specific antigens are proteins or other molecules that are present on melanoma cells but not normally found on healthy cells in the body. These antigens can be recognized by the immune system as foreign and trigger an immune response, making them potential targets for immunotherapy treatments for melanoma.

There are two main types of melanoma-specific antigens: tumor-specific antigens (TSAs) and tumor-associated antigens (TAAs). TSAs are unique to cancer cells and are not found on normal cells, while TAAs are overexpressed or mutated versions of proteins that are also present in normal cells.

Examples of melanoma-specific antigens include Melan-A/MART-1, gp100, and tyrosinase. These antigens have been studied extensively as targets for cancer vaccines, adoptive cell therapy, and other immunotherapy approaches to treat melanoma.

An allele is a variant form of a gene that is located at a specific position on a specific chromosome. Alleles are alternative forms of the same gene that arise by mutation and are found at the same locus or position on homologous chromosomes.

Each person typically inherits two copies of each gene, one from each parent. If the two alleles are identical, a person is said to be homozygous for that trait. If the alleles are different, the person is heterozygous.

For example, the ABO blood group system has three alleles, A, B, and O, which determine a person's blood type. If a person inherits two A alleles, they will have type A blood; if they inherit one A and one B allele, they will have type AB blood; if they inherit two B alleles, they will have type B blood; and if they inherit two O alleles, they will have type O blood.

Alleles can also influence traits such as eye color, hair color, height, and other physical characteristics. Some alleles are dominant, meaning that only one copy of the allele is needed to express the trait, while others are recessive, meaning that two copies of the allele are needed to express the trait.

An AIDS vaccine is a type of preventive vaccine that aims to stimulate the immune system to produce an effective response against the human immunodeficiency virus (HIV), which causes acquired immunodeficiency syndrome (AIDS). The goal of an AIDS vaccine is to induce the production of immune cells and proteins that can recognize and eliminate HIV-infected cells, thereby preventing the establishment of a persistent infection.

Despite decades of research, there is still no licensed AIDS vaccine available. This is due in part to the unique challenges posed by HIV, which has a high mutation rate and can rapidly evolve to evade the immune system's defenses. However, several promising vaccine candidates are currently being tested in clinical trials around the world, and researchers continue to explore new approaches and strategies for developing an effective AIDS vaccine.

Interleukin-17 (IL-17) is a type of cytokine, which are proteins that play a crucial role in cell signaling and communication during the immune response. IL-17 is primarily produced by a subset of T helper cells called Th17 cells, although other cell types like neutrophils, mast cells, natural killer cells, and innate lymphoid cells can also produce it.

IL-17 has several functions in the immune system, including:

1. Promoting inflammation: IL-17 stimulates the production of various proinflammatory cytokines, chemokines, and enzymes from different cell types, leading to the recruitment of immune cells like neutrophils to the site of infection or injury.
2. Defending against extracellular pathogens: IL-17 plays a critical role in protecting the body against bacterial and fungal infections by enhancing the recruitment and activation of neutrophils, which can engulf and destroy these microorganisms.
3. Regulating tissue homeostasis: IL-17 helps maintain the balance between immune tolerance and immunity in various tissues by regulating the survival, proliferation, and differentiation of epithelial cells, fibroblasts, and other structural components.

However, dysregulated IL-17 production or signaling has been implicated in several inflammatory and autoimmune diseases, such as psoriasis, rheumatoid arthritis, multiple sclerosis, and inflammatory bowel disease. Therefore, targeting the IL-17 pathway with specific therapeutics has emerged as a promising strategy for treating these conditions.

Lymphatic diseases refer to a group of conditions that affect the lymphatic system, which is an important part of the immune and circulatory systems. The lymphatic system consists of a network of vessels, organs, and tissues that help to transport lymph fluid throughout the body, fight infection, and remove waste products.

Lymphatic diseases can be caused by various factors, including genetics, infections, cancer, and autoimmune disorders. Some common types of lymphatic diseases include:

1. Lymphedema: A condition that causes swelling in the arms or legs due to a blockage or damage in the lymphatic vessels.
2. Lymphoma: A type of cancer that affects the lymphatic system, including Hodgkin's and non-Hodgkin's lymphoma.
3. Infections: Certain bacterial and viral infections can affect the lymphatic system, such as tuberculosis, cat-scratch disease, and HIV/AIDS.
4. Autoimmune disorders: Conditions such as rheumatoid arthritis, lupus, and scleroderma can cause inflammation and damage to the lymphatic system.
5. Congenital abnormalities: Some people are born with abnormalities in their lymphatic system, such as malformations or missing lymph nodes.

Symptoms of lymphatic diseases may vary depending on the specific condition and its severity. Treatment options may include medication, physical therapy, surgery, or radiation therapy. It is important to seek medical attention if you experience symptoms of a lymphatic disease, as early diagnosis and treatment can improve outcomes.

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.

Epitope mapping is a technique used in immunology to identify the specific portion or regions (called epitopes) on an antigen that are recognized and bind to antibodies or T-cell receptors. This process helps to understand the molecular basis of immune responses against various pathogens, allergens, or transplanted tissues.

Epitope mapping can be performed using different methods such as:

1. Peptide scanning: In this method, a series of overlapping peptides spanning the entire length of the antigen are synthesized and tested for their ability to bind to antibodies or T-cell receptors. The peptide that shows binding is considered to contain the epitope.
2. Site-directed mutagenesis: In this approach, specific amino acids within the antigen are altered, and the modified antigens are tested for their ability to bind to antibodies or T-cell receptors. This helps in identifying the critical residues within the epitope.
3. X-ray crystallography and NMR spectroscopy: These techniques provide detailed information about the three-dimensional structure of antigen-antibody complexes, allowing for accurate identification of epitopes at an atomic level.

The results from epitope mapping can be useful in various applications, including vaccine design, diagnostic test development, and understanding the basis of autoimmune diseases.

Lysosome-Associated Membrane Protein 1 (LAMP-1) is a type I transmembrane protein that is heavily glycosylated and primarily localized to the limiting membrane of lysosomes. It is one of the most abundant proteins in the lysosomal membrane, making up approximately 50% of its total protein mass. LAMP-1 plays a crucial role in maintaining the integrity and stability of the lysosomal membrane by preventing lysosomal enzyme leakage into the cytosol. It also participates in various cellular processes, including autophagy, cell death, and antigen presentation.

LAMP-1 is often used as a marker for late endosomes and lysosomes due to its specific localization in these organelles. The protein contains several structural features that are important for its function, such as a large luminal domain with multiple glycosylation sites, a transmembrane domain, and a short cytoplasmic tail. The cytoplasmic tail interacts with various proteins involved in intracellular trafficking, membrane fusion, and cytoskeletal organization, which contributes to the proper functioning of lysosomes and other related organelles.

ZAP-70 (zeta-associated protein-70) is a protein tyrosine kinase that plays a critical role in T-cell antigen receptor (TCR) signal transduction. It is primarily expressed in T-cells and natural killer cells. Upon TCR engagement, ZAP-70 becomes activated and phosphorylates downstream signaling molecules, leading to the activation of various cellular responses such as cytokine production, proliferation, differentiation, and survival.

Defects in ZAP-70 function have been implicated in various immune disorders, including severe combined immunodeficiency (SCID) and autoimmune diseases. Mutations in the ZAP-70 gene can lead to impaired T-cell activation and differentiation, resulting in immunodeficiency. On the other hand, overactivation of ZAP-70 has been associated with the development of autoimmunity. Therefore, maintaining appropriate regulation of ZAP-70 activity is essential for normal immune function.

The Immunoglobulin (Ig) variable region is the antigen-binding part of an antibody, which is highly variable in its amino acid sequence and therefore specific to a particular epitope (the site on an antigen that is recognized by the antigen-binding site of an antibody). This variability is generated during the process of V(D)J recombination in the maturation of B cells, allowing for a diverse repertoire of antibodies to be produced and recognizing a wide range of potential pathogens.

The variable region is composed of several sub-regions including:

1. The heavy chain variable region (VH)
2. The light chain variable region (VL)
3. The heavy chain joining region (JH)
4. The light chain joining region (JL)

These regions are further divided into framework regions and complementarity-determining regions (CDRs). The CDRs, particularly CDR3, contain the most variability and are primarily responsible for antigen recognition.

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.

Distinguishing between human lymphocyte subsets. Identification of cone cell inner and outer segments and to a lesser extent ...
May 2017). "Essential Roles of SATB1 in Specifying T Lymphocyte Subsets". Cell Reports. 19 (6): 1176-1188. doi:10.1016/j.celrep ... SATB1-deficient thymocytes enter inappropriate T lineages and fail to generate the NKT and Treg subsets. The Treg deficiency ...
August 2020). "Lymphocyte Subset Counts in COVID-19 Patients: A Meta-Analysis". Cytometry. Part A. 97 (8): 772-776. doi:10.1002 ... Sallusto F, Lenig D, Förster R, Lipp M, Lanzavecchia A (October 1999). "Two subsets of memory T lymphocytes with distinct ... revealed that another T helper subset may exist. Th9 cells are claimed to be an IL9 (interleukin 9)-producing T cell subset ... These effects are primarily due to the loss of any helper T cell that can interact with the B lymphocyte correctly. Another ...
September 2020). "Dynamic changes in peripheral blood lymphocyte subsets in adult patients with COVID-19". International ... It is well known that a SARS-CoV-2 infection induces a decreased lymphocyte count, and those with a lower lymphocyte count ... The authors pointed out that the lymphocyte count recovered to normal after 4 days. Low lymphocyte count could be a significant ... Zhang HJ, Qi GQ, Gu X, Zhang XY, Fang YF, Jiang H, Zhao YJ (July 2021). "Lymphocyte blood levels that remain low can predict ...
el-Demiry M, James K (1988). "Lymphocyte subsets and macrophages in the male genital tract in health and disease. A monoclonal ... effects of activin and transforming growth factor beta on lymphocyte subsets in vitro". Biology of Reproduction. 58 (4): 943- ... T-lymphocytes (T-cells) are white blood cells which take part in cell-mediated immunity. They are often found within tissues ... B-lymphocytes take part in the adaptive immune response and produce antibodies. These cells are not normally found in the ...
Poussier, P., et al., Lymphopenia and abnormal lymphocyte subsets in the "BB" rat: relationship to the diabetic syndrome. ...
"Transitional Type 1 and 2 B Lymphocyte Subsets Are Differentially Responsive to Antigen Receptor Signaling". J. Biol. Chem. 277 ... Chung JB, Sater RA, Fields ML, Erikson J, Monroe JG (2002). "CD23 defines two distinct subsets of immature B cells which differ ... T1 B cells are distinguished from the other subsets by the following surface marker characteristics: they are IgMhiIgD−CD21− ... B cells, Lymphocytes, Bone marrow, Human cells, Immunology, Immune system). ...
He was among those who defined the subsets of human B lymphocytes. These studies led to a redefinition of many human B cell ... "Analysis of somatic mutation in five B cell subsets of human tonsil". The Journal of Experimental Medicine. 180 (1): 329-339. ... "Receptor revision of immunoglobulin heavy chain variable region genes in normal human B lymphocytes". The Journal of ...
1996;17(2):197-9. PMID 8936281 Differences in intraepithelial lymphocyte t cell subsets isolated from murine small versus large ... his research was on experimental models of hapten-induced Inflammatory bowel disease and studying intraepithelial lymphocytes. ...
Sallusto, F.; Lenig, D.; Förster, R.; Lipp, M.; Lanzavecchia, A. (1999). "Two subsets of memory T lymphocytes with distinct ... Viola, A.; Schroeder, S.; Sakakibara, S.; Lanzavecchia, A. (1999). "T lymphocyte costimulation mediated by reorganization of ... lymphocyte activation and immunological memory and iii) human monoclonal antibodies. In 1985, using antigen-specific T and B ... and discovered a fundamental division of memory T cells into two major subsets of central memory and effector memory and ...
885-895 Han S, Dillon SR... Kelsoe G (1997). V(D)J recombinase activity in a subset of germinal center B lymphocytes. Science. ... Kelsoe's research focuses on humoral immunity, B lymphocyte development and activation, and the mechanism by which lymphocytes ...
"Two subsets of memory T lymphocytes with distinct homing potentials and effector functions." Nature 401.6754 (1999): 708. ...
Sallusto F, Lenig D, Förster R, Lipp M, Lanzavecchia A (1999). "Two subsets of memory T lymphocytes with distinct homing ... These two subsets were previously called "naturally occurring" and "adaptive" (or "induced"), respectively. Both subsets ... Gamma delta T cells (γδ T cells) represent a small subset of T cells which possess a γδ TCR rather than the αβ TCR on the cell ... T cell subsets were initially defined by function, but also have associated gene or protein expression patterns. T helper cells ...
... s are a subset of T lymphocytes that might have some of the same functions as memory B cells. Their lineage is ... Sallusto F, Lenig D, Förster R, Lipp M, Lanzavecchia A (October 1999). "Two subsets of memory T lymphocytes with distinct ... Those lymphocytes are capable of self-renewal as are the TCM lymphocytes and are also capable of generating both the TCM and ... subsets. Although most information is currently based on observations in the cytotoxic T cells (CD8-positive) subset, similar ...
This selective inhibition is not observed in conventional chemotherapy which reduces the number of all lymphocyte subsets. In ...
"Distribution of cyclophilin B-binding sites in the subsets of human peripheral blood lymphocytes". Immunology. 91 (4): 609-17. ... This protein can bind to cells derived from T- and B-lymphocytes, and may regulate cyclosporine A-mediated immunosuppression. ... recognized by HLA-A24-restricted and tumor-specific cytotoxic T lymphocytes which could be used as cancer vaccines, and in fact ...
Effects of Lycium barbarum polysaccharide on tumor microenvironment T-lymphocyte subsets and dendritic cells in H22-bearing ...
Harris MT, Schwarting GA, Stout RD (September 1981). "Selective expression of asialo GM1 on maturational subsets of lymphocytes ... 15 July 1997). "Functional and phenotypic characterization of cord blood and bone marrow subsets expressing FLT3 (CD135) ... in vivo differentiation into lymphocytes and potential for germ line transmission". FEBS Letters. 455 (1-2): 101-4. doi:10.1016 ...
In jawless fishes, two subsets of lymphocytes use variable lymphocyte receptors (VLRs) for antigen binding. Diversity is ... T and B lymphocytes are the cells of the adaptive immune system. The human body has about 2 trillion lymphocytes, which are 20- ... Lymphocyte receptors, Ig and TCR, are found in all jawed vertebrates. The most ancient Ig class, IgM, is membrane-bound and ... Cytotoxic T cells (also known as TC, killer T cell, or cytotoxic T-lymphocyte (CTL)) are a sub-group of T cells that induce the ...
There are no major changes in the T-lymphocytes subsets of the mesenterial lymph node. Instead γδ-T cells guide the immune ... Antigens of C. oncophora larvae and adult worms are capable of triggering lymphocyte proliferation. Moreover, excretory/ ... Primary infection does not involve recruitment of specific lymphocytes to the intestinal mucosa. ...
PTSAgs induce the VB-specific expansion of both CD4 and CD8- subsets of T-lymphocytes. TSST-1 forms homodimers in most of its ...
"Specific lymphocyte subsets predict response to adoptive cell therapy using expanded autologous tumor-infiltrating lymphocytes ... Tumor infiltrating lymphocyte entry in the public domain NCI Dictionary of Cancer Terms Lion Biotechnologies, Inc. (Lymphocytes ... CD8+-Enriched Autologous Tumor-infiltrating Lymphocytes Following a Lymphocyte Depleting Regimen in Metastatic Digestive Tract ... Tumor-infiltrating lymphocytes (TIL) are white blood cells that have left the bloodstream and migrated towards a tumor. They ...
2001a;97:319-327 Lymphocyte phenotype subsets in the cerebrospinal fluid of normal horses and horses with equine protozoal ...
"The Monoclonal Antimonocyte Antibody My4 Stains B Lymphocytes and Two Distinct Monocyte Subsets in Human Peripheral Blood". ... The first clear description of monocyte subsets by flow cytometry dates back to the late 1980s, when a population of CD16- ... Analysis of monocyte subsets has demonstrated predominance of classical monocytes and absence of CD14lowCD16+ monocytes. The ... Both may be useful, but these cells became valid diagnostic tools only when monocyte subsets are determined. Monocytic cells ...
Knockout mice that lack TOX have a severe defect in development of certain subsets of T lymphocytes. TOX is necessary for T ... Patients with cancer typically have high levels of TOX in their tumor-infiltrating lymphocytes, and anti-tumor immunity is ... TOX is highly expressed in the thymus, the site of development of T lymphocytes. ...
Berrington JE, Barge D, Fenton AC, Cant AJ, Spickett GP (May 2005). "Lymphocyte subsets in term and significantly preterm UK ... The two main types of lymphocytes are B lymphocytes and T lymphocytes. B lymphocytes make antibodies, and T lymphocytes help ... A lymphocyte is a type of white blood cell (leukocyte) in the immune system of most vertebrates. Lymphocytes include T cells ( ... A lymphocyte count is usually part of a peripheral complete blood cell count and is expressed as the percentage of lymphocytes ...
Nassar GM, Montero A, Fukunaga M, Badr KF (1997). "Contrasting effects of proinflammatory and T-helper lymphocyte subset-2 ... and B-lymphocytes express ALOX5. Platelets, T cells, and erythrocytes are ALOX5-negative. In skin, Langerhans cells strongly ... "Studies on the regulation and localization of 5-lipoxygenase in human B-lymphocytes". Eur. J. Biochem. 232 (1): 37-46. doi: ...
... differential signaling requirements for activation of assembled cyclin D3-cdk4 complexes in B-1 and B-2 lymphocyte subsets". ...
Ademmer; Ebert; Müller-Ostermeyer; Friess; Büchler; Schubert; Malfertheiner (April 1998). "Effector T lymphocyte subsets in ... 2006). "Profiling lymphocyte subpopulations in peripheral blood under efalizumab treatment of psoriasis by multi epitope ligand ... "Automatic Recognition of Muscle-Invasive T-Lymphocytes Expressing Dipeptidyl-Peptidase IV (CD26) and Analysis of the Associated ... "Poisson Numbers and Poisson Distributions in Subset Surprisology". Annals of Combinatorics. 8 (4): 473-485. doi:10.1007/s00026- ...
"In Vitro Measles Virus Infection of Human Lymphocyte Subsets Demonstrates High Susceptibility and Permissiveness of both Naive ... In the later stages of infection, the virus infects other immune cell types, including B cells and T lymphocytes also via ... Three receptors for the H protein have been identified to date: complement regulatory molecule CD46, the signaling lymphocyte ...
... these specific transcription factors activate or repress target genes critical in the differentiation of the lymphocyte subsets ... Studies have confirmed the ability of some ILC subsets to convert into a different subset in the presence of specific cytokines ... There are two subsets of ILC3s, NCR- and NCR+ ILC3s, with the displayed NCR on mice ILC3s being NKp46, in comparison to NKp44 ... All ILC subsets are present in the liver and regulate the immune response to protect the tissue from viral and bacterial ...
  • Levels of lymphocyte subsets were determined by flow cytometry. (providence.org)
  • Also, whole blood was recovered, and the number of circulating total leukocytes, as well as specific lymphocyte subsets, was determined by flow cytometry. (cdc.gov)
  • Clinical changes in disease expression measured by a disease activity score were correlated with changes in percentages of peripheral blood lymphocyte subsets. (northwestern.edu)
  • Eisenstein, DM, O'Gorman, MRG & Pachman, LM 1997, ' Correlations between change in disease activity and changes in peripheral blood lymphocyte subsets in patients with juvenile dermatomyositis ', Journal of Rheumatology , vol. 24, no. 9, pp. 1830-1832. (northwestern.edu)
  • Production of interferons and change of the lymphocyte subpopulation phenotype in peripheral blood at cervical papillomavirus infection. (wikigenes.org)
  • 001). The peripheral CD8+ lymphocyte subpopulation includes four distinct cell subsets: TCRgammadeltaCD8alphaalpha T cells, CD3 --CD8alphaalpha lymphocytes (NK cells) and TCRalphabetaCD8alphabeta T cells, and TCRalphabetaCD4+CD8alphaalpha, and putatively TCRalphabetaCD4 --CD8alphaalpha T cells. (iastate.edu)
  • Our data demonstrate that ZAP-70 is expressed in a subpopulation of tonsillar and splenic normal B-lymphocytes that express an activated phenotype. (elsevierpure.com)
  • The heterogeneity of tumor infiltrating lymphocytes (TILs) is not well characterized in brain metastasis. (akoyabio.com)
  • Comparison of tumor biopsies before and after treatment reveals an increase of activated, conventional CD4+ tumor-infiltrating lymphocytes (TIL) in most patients and higher clonality by TCRβ sequencing. (bepress.com)
  • Researchers determined that the number of tumor-infiltrating lymphocytes (TILs) present in esophageal cancer tumors correlated with patient long-term survival rates. (sciencedaily.com)
  • A subset of these are called tumor-infiltrating lymphocytes (TILs). (sciencedaily.com)
  • Altered cellular immune function has been demonstrated in patients with lung cancer, including decreased numbers of circulating lymphocytes and changes in the percentage of lymphocytes in various functional subsets. (elsevierpure.com)
  • CD45 lymphocyte count was designated the total lymphocyte count. (cdc.gov)
  • Lymphocyte populations in cancer patients were compared to those of nonsmokers and a smoking patient population. (elsevierpure.com)
  • To investigate the effects of age and gender on human lymphocyte populations, we studied lymphocyte subsets and their expression of activation markers in peripheral blood in healthy people above the age of 21, and analyzed this according to gender. (biomedcentral.com)
  • Evidence from this study indicates that pulmonary exposure to certain welding fumes cause decrements in systemic immune cell populations, specifically circulating T-lymphocytes, and these alterations in immune cell number are not dependent exclusively on Mn, but likely a combination of other metals present in welding fume. (cdc.gov)
  • Breg cell, regulatory B cell T lymphocyte The percentage of CD4+ T cells at different stages of differentiation were calculated, and significant differences between the AS patients and HCs are shown in Fig.?2. (scienza-under-18.org)
  • B10 cell, IL-10 producing regulatory B cell The number of regulatory lymphocytes detected in the blood of the AS patients changed significantly after Anbainuo treatment, with the percentage of Treg FLJ20315 cells (CD3+CD4+CD25+CD127? (scienza-under-18.org)
  • The cause of the change in percentage of lymphocyte subsets with age, and the different effects on males and females is not fully understood but warrants further study. (biomedcentral.com)
  • Changes in the percentage of peripheral blood CD 19 positive lymphocytes (B cells) correlated with changes in disease activity (Spearman rank coefficients = 0.47, p = 0.02). (northwestern.edu)
  • With CD45 gating, the relative numbers of beads and lymphocyte subsets are enumerated, and their absolute numbers and percentage values are calculated. (cdc.gov)
  • Single-platform technology (SPT) is designed to enable de- system and managing the health care of persons infected with terminations of both absolute and percentage lymphocyte sub- human immunodeficiency virus (HIV) ( 1-4 ). (cdc.gov)
  • SH2D1A was positively correlated with immunoglobulin A , immunoglobulin G and TL subsets (CD3+, CD4+ and CD8+) (P (bvsalud.org)
  • Enumeration of CD4+ lymphocytes in HIV-positive participants and age-matched controls was performed on cryopreserved whole blood using the method reported by Fiebig et. (cdc.gov)
  • De Paoli P, Reitano M, Battistin S, Castiglia C, Santini G. Enumeration of human lymphocyte subsets by monoclonal antibodies and flow cytometry: a comparative study using whole blood or mononuclear cells separated by density gradient centrifugation. (bdbiosciences.com)
  • We hypothesized that CLA modifies peripheral lymphocyte phenotype, regardless of the environment. (iastate.edu)
  • Owing to the prognostic value of B-CLL ZAP-70 expression, this phenotype may reflect intrinsic biological differences between the two subsets of disease. (elsevierpure.com)
  • The expression of three of these genes was further tested by quantitative RT-PCR in lymphocyte subpopulations [CD4+ and CD8+ T, CD19+ B, CD56+ natural killer cells and peripheral blood lymphocytes (PBLs)] from ten adult donors. (sckcen.be)
  • The level of BAX activation in the various subpopulations correlated with the sensitivity of the cells to radiation, suggesting its possible role in the differential radiosensitivity of hematopoietic cell subsets. (sckcen.be)
  • In this study we have established culture conditions that allow the preferential and rapid expansion of either T cell receptor (TCR) + /CD3 + 16 - T lymphocytes or TCR - /CD3 - 16 - natural killer (NK) cells, or the non-selective outgrowth of both subsets. (wustl.edu)
  • In seven patients (44%), the abnormal ratio resulted from an increase in the absolute number of suppressor cells with a normal number of helper T lymphocytes. (elsevierpure.com)
  • We quantitated the subsets of immune-cells utilizing a targeted panel of proteins including PanCK, CD8, CD4, VISTA and IBA-1, and analyzed an average of 15,000 cells per sample. (akoyabio.com)
  • However, we have identified a distinct subset of human NK cells that are deficient for FcRγ expression but express normal levels of CD3ζ. (nih.gov)
  • Thus, our study reveals FcRγ-deficient NK cells as a novel subset of human NK cells that have remarkably potent responses toward antibody-coated targets. (nih.gov)
  • Various subsets of invariant natural killer T (iNKT) cells with different cytokine productions develop in the mouse thymus, but the factors driving their differentiation remain unclear. (nih.gov)
  • Furthermore, MP increased WBC and HGB counts and reduced the ratio of G0/G1 phase obviously, increased the proportion of S phase and G2/M phase in peripheral blood lymphocytes, increased the counts of CD4+ T cells and the ratio of CD4+/CD8+, and reduced the inhibition rate of splenic lymphocytes. (hindawi.com)
  • Recent studies using neutralizing anti-CCL5 antibodies have suggested that the constitutive expression of CCL5 plays a regulatory role in maintaining tissue-resident memory (Trm) T cells in the human vaginal tract 9 and mouse skin 10 , suggesting that constitutive CCL5 expression in noninflamed conditions, hereafter referred to as homeostatic CCL5 expression, might be important for homeostasis of tissue-resident lymphocytes. (nature.com)
  • However, it remains unclear whether CLL-B cells aberrantly acquire ZAP-70 expression during the transformation process or whether ZAP-70 may be expressed under certain conditions in normal human B-lymphocytes. (elsevierpure.com)
  • CD27, a lymphocyte-specific member of the TNF/NGF-R family, is expressed on the majority of peripheral blood T cells. (aai.org)
  • There is evidence that also in vivo, CD4+ cells that have persistently been stimulated by Ag, accumulate within the CD45RA-CD27- subset. (aai.org)
  • In agreement with earlier findings, all CD4+CD45RA+CD45RO- T cells were found to express CD27, whereas a small fraction of the CD4+CD45RA-CD45RO+ subset lacks the molecule. (aai.org)
  • CD4+ T-cells and other lymphocyte subsets in persons infected with human immunodeficiency virus (HIV). (cdc.gov)
  • HIV infection is characterized by a decrease and, eventually, a depletion of CD4+ T-lymphocytes (helper T cells). (cdc.gov)
  • Using immunophenotyping, HIV-positive blood samples and age-matched controls were tested for the proportion of lymphocytes that are T cells, B cells, natural killer (NK) cells, CD4+ T cells (helper T cells), and CD8+ T cells (suppressor/inducer T cells). (cdc.gov)
  • Mycobacterium bovis, the causative agent of bovine tuberculosis is known to persist within granulomas, distinct lesions represented by a caseonecrotic core surrounded by epithelioid macrophages, T-lymphocytes, B-lymphocytes, Langhans-type multinucleated giant cells and fibroblasts. (usda.gov)
  • and (iii) the dramatic reductions to lymphocyte numbers and function 1-2 h after exercise reflects a transient and time-dependent redistribution of immune cells to peripheral tissues, resulting in a heightened state of immune surveillance and immune regulation, as opposed to immune suppression. (frontiersin.org)
  • about 75% of the lymphocytes are T cells, 20% B cells, and 5% NK cells. (msdmanuals.com)
  • Thus, this study investigated the effects of masturbation-induced orgasm on lymphocyte circulation and cytokine production in healthy young males. (karger.com)
  • Limitations: Although elevations of lymphocyte subsets in major depressive and OCD patients were observed, conclusions concerning immune status in OCD ought to be held in abeyance pending assessment of other indices of immune and cytokine functioning. (aku.edu)
  • Monocyte subsets display age-dependent alterations at fasting and undergo non-age-dependent changes following consumption of a meal. (usda.gov)
  • In addition, leukocyte and lymphocyte subsets were analyzed via flow cytometry, and the production of lipopolysaccharide-induced interleukin 6 and tumor necrosis factor alpha was measured before and then 5 and 45 min after the orgasm. (karger.com)
  • The elements were evaluated by the total number of peripheral leukocyte, granulocyte and lymphocyte ratio and lymphocyte subsets. (scirp.org)
  • The number of leukocyte subsets, granulocyte and lymphocyte were also regulated by dose dependent manner. (scirp.org)
  • Optimal proliferation of lymphocytes was obtained using a combination of irradiated allogeneic peripheral blood lymphocytes (PBL) and irradiated Epstein Barr virus (EBV) transformed lymphoblastoid B cell lines (B-LCL). (wustl.edu)
  • Cumulative physiological stress is associated with age-related changes to peripheral T lymphocyte subsets in healthy humans. (usda.gov)
  • During the initial phase of the disease, patients demonstrated an increase in the total lymphocyte counts (CD3). (tubitak.gov.tr)
  • The guidelines describe single-platform technology (SPT), a process in which absolute counts of lymphocyte subsets are measured from a single tube by a single instrument. (cdc.gov)
  • T-lymphocyte counts, with special emphasis and stunted children has not changed dra- on the effect of nutritional rehabilitation. (who.int)
  • Serum from145 iGCTs patients were collected and lymphocyte subsets were analyzed by flow cytometry. (researchsquare.com)
  • Lymphocyte subset analysis by flow cytometry. (bdbiosciences.com)
  • Clinical Applications of Flow Cytometry: Quality Assurance and Immunophenotyping of Lymphocytes: Approved Guideline. (bdbiosciences.com)
  • Lymphocyte immunophenotyping by flow cytometry in normal adults: comparison of fresh whole blood lysis technique, Ficoll-Paque separation and cryopreservation. (bdbiosciences.com)
  • Human lymphocyte subpopulations differ in their cellular responses to ionizing radiation. (sckcen.be)
  • In contrast to DDB2, TNFRSF10B and BAX were differentially modulated among the subpopulations and the PBLs, being more activated in irradiated CD19+ B and CD8+ T lymphocytes. (sckcen.be)
  • In contrast, within the CD8+ compartment CD27- subsets were found both in the CD45RA+ and CD45RA- subpopulations. (aai.org)
  • If the complete blood count reveals lymphocytopenia, testing for immunodeficiency and analysis of lymphocyte subpopulations should follow, usually after the patient has recovered from any acute event. (msdmanuals.com)
  • This study retrospectively analyzed serum lymphocyte subsets and survival outcomes in intracranial germ cell tumors (iGCTs) patients. (researchsquare.com)
  • The levels of SH2D1A, signaling lymphocyte activation molecule (SLAM) and SLAM-associated protein (SAP) were significantly higher in the IM group than those in the control and CAEBV groups (P (bvsalud.org)
  • Identification of a molecule uniquely expressed on a gamma/delta TCR+ subset within bovine intestinal intraepithelial lymphocytes. (ox.ac.uk)
  • Because lymphocytes account for only 20 to 40% of the total white blood cell (WBC) count, lymphocytopenia may go unnoticed if the WBC count is checked without a differential. (msdmanuals.com)
  • polymorphonuclear leukocytes, and neutrophil ratios increased and eosinophil, mononuclear leukocytes, lymphocyte, and monocyte ratios were decrease when these values were compared to changes between days. (yyu.edu.tr)
  • It is common to evaluate lymphocytes subsets in virus infection, immunodeficiency diseases and cancer. (researchsquare.com)
  • Human Immunodeficiency Virus (HIV) Infection Human immunodeficiency virus (HIV) infection results from 1 of 2 similar retroviruses (HIV-1 and HIV-2) that destroy CD4+ lymphocytes and impair cell-mediated immunity, increasing risk of certain. (msdmanuals.com)
  • Effect of Graves' disease intrathyroidal lymphocytes (ITL) on xenotransplants of human thyroid tissue in athymic nude mice. (wikigenes.org)
  • Islet-infiltrating lymphocytes of individual male and female non-obese diabetic (NOD) mice were examined with the purpose of determining the differences that lead to a predominance of diabetes in female versus males NOD mice. (arizona.edu)
  • When normalized for the amount of islet lymphocytes recovered, the infiltrating lymphocytes of female NOD mice were indistinguishable from those of male NOD mice. (arizona.edu)
  • STUDY DESIGN AND METHODS: The hematopoietic progenitor cell (HPC) and lymphocyte subset (LS) content of approximately 8000 CB units were characterized, and these results were correlated with donor ethnicity, birth weight, gestational age, sex, and type of delivery. (emmes.com)
  • Lesions at all time-points contained large numbers of CD4+ T-lymphocytes with lesser numbers of CD8+ T-lymphocytes and few gamma/delta T-lymphocytes. (usda.gov)
  • Distinguishing between human lymphocyte subsets. (wikipedia.org)
  • To discriminate between these two possibilities, we assessed ZAP-70 expression in normal human B-lymphocytes. (elsevierpure.com)
  • A low ratio of CD4+ (helper) any chromosomal or hereditary disorder lymphocytes relative to CD8+ (suppressor) that caused the malnutrition. (who.int)
  • We quantitated lymphocyte subsets in 54 patients with lung cancer including patients with limited (stages 1 and 2) nonsmall cell lung cancer (NSCLC, n = 23), advanced (stage 3) NSCLC (n = 16), and small cell cancer (SCLC, n = 15). (elsevierpure.com)
  • Serum albumin was decreased in 15 lung cancer patients, and lymphocyte subsets were separately evaluated in these patients. (elsevierpure.com)
  • A decrease in population of T-lymphocytes subsets is frequent in patients with SCLC, advanced NSCLC, and lung cancer patients with hypoalbuminemia. (elsevierpure.com)
  • Method: Circulating lymphocytes were determined from morning blood samples taken from OCD and major depressive patients, as well as from age- and sex-matched controls. (aku.edu)
  • Finally, in a subset of patients, symptoms of the illness, stress/coping, and circulating lymphocytes, were also evaluated following 12 weeks of antidepressant medication (serotonergic reuptake inhibitor). (aku.edu)
  • To investigate whether changes in peripheral blood lymphocytes correlate with changes in disease activity in juvenile dermatomyositis (JDM). (northwestern.edu)
  • It is important to note that the lymphocytes in the blood represent only a small proportion of the total lymphocyte pool, and the lymphocyte count may not always correlate with the composition and numbers of lymphocytes in other lymphoid (eg, nodes, spleen) and non-lymphoid (eg, lung, liver) tissues. (msdmanuals.com)
  • Kidd P, Vogt R. Report of the workshop on the evaluation of T-cell subsets during HIV infection and AIDS. (bdbiosciences.com)
  • In acute viremia due to HIV or other viruses, lymphocytes may undergo accelerated destruction from active infection with the virus, may be trapped in the spleen or lymph nodes, or may migrate to the respiratory tract. (msdmanuals.com)
  • All enrolled thymic lymphocytes is widely accepted as infants were breastfed in addition to receiv- an indicator of the depression of thymus- ing some traditional foods, according to dependent immune competence associated their age. (who.int)
  • Lymphocytopenia may also reflect impaired lymphocyte production arising from destruction of thymic or lymphoid architecture. (msdmanuals.com)
  • Circulating activated lymphocyte subsets as potential blood biomarkers of cancer progression. (providence.org)
  • The objective of this study was to predict the value of lymphocyte subsets in cancer progression. (providence.org)
  • CLA synergized with a Brachyspira hyodysenteriae bacterin vaccine to expand the TCRgammadeltaCD8alphaalpha and NK cell subsets. (iastate.edu)
  • To address this, we performed a targeted analysis of immune-cell subsets in brain metastasis tissues to test immunosuppressive routes involved in brain metastasis. (akoyabio.com)
  • There were no significant correlations in disease activity with changes of T cell subsets or the T cell activation markers CD25 or DR. Conclusion. (northwestern.edu)
  • The mechanism was perhaps related to inhibiting lymphocyte apoptosis and promoting the balance of Th1/Th2 cell subsets. (hindawi.com)
  • This study was undertaken to analyze the regulation of CD27 on different T-cell subsets and to determine whether the loss of CD27 expression is an irreversible event and may thereby mark T-cell differentiation. (aai.org)
  • The Cambridge Biotech HIV-1 Western Blot Kit is manufactured by Calypte Corporation from HIV-I propagated in an H9/HTLV-IIIb T-lymphocyte cell line. (cdc.gov)
  • Influence of racial background on the distribution of T-cell subsets and Leu 11-positive lymphocytes in healthy blood donors. (bdbiosciences.com)
  • Analysis of T cell subsets in normal adults. (bdbiosciences.com)
  • McNerlan, SE, Alexander, HD & Rea, IM 1999, ' Age-related reference intervals for lymphocyte subsets in whole blood of healthy individuals ', Scandinavian Journal of Clinical and Laboratory Investigation , vol. 59, no. 2, pp. 89-92. (ulster.ac.uk)
  • Circulating activated lymphocyte subsets as potential blood biomarkers" by Ying-Yi Wang, Na Zhou et al. (providence.org)
  • The secondary objective was to assess the effect of anti-OX40 on lymphocyte subsets in the tumor and blood. (bepress.com)
  • The welding fume with the lower concentration of Mn had no significant effect on the numbers of blood lymphocytes and lymphocyte subsets compared to control values. (cdc.gov)
  • The level of The infants with PEM were enrolled in T-lymphocyte subsets in peripheral blood the study after fulfilling a set of inclusion provide information about the development criteria. (who.int)
  • To understand the correlation between changes in disease status (including CRP, BASDAI, and ASDAS) and changes in lymphocyte frequency after Anbainuo therapy, Spearmans rank correlation analyses showed that the decrease in CRP was positively correlated with the increase in the frequency of Tregs (CD3+CD4+CD25+CD127? (scienza-under-18.org)
  • Dynamic monitoring of serum lymphocyte subsets can be used as an auxiliary indicator for prognosis judgment. (researchsquare.com)
  • To more fully phenotypically characterize the lymphocyte subsets that are most influenced by dietary CLA supplementation and to elucidate the functional implications of their expansion, we conducted the second experiment. (iastate.edu)
  • Regulation of CD27 expression on subsets of mature T-lymphocytes. (aai.org)