A glycoprotein that is central in both the classical and the alternative pathway of COMPLEMENT ACTIVATION. C3 can be cleaved into COMPLEMENT C3A and COMPLEMENT C3B, spontaneously at low level or by C3 CONVERTASE at high level. The smaller fragment C3a is an ANAPHYLATOXIN and mediator of local inflammatory process. The larger fragment C3b binds with C3 convertase to form C5 convertase.
Complement activation initiated by the interaction of microbial ANTIGENS with COMPLEMENT C3B. When COMPLEMENT FACTOR B binds to the membrane-bound C3b, COMPLEMENT FACTOR D cleaves it to form alternative C3 CONVERTASE (C3BBB) which, stabilized by COMPLEMENT FACTOR P, is able to cleave multiple COMPLEMENT C3 to form alternative C5 CONVERTASE (C3BBB3B) leading to cleavage of COMPLEMENT C5 and the assembly of COMPLEMENT MEMBRANE ATTACK COMPLEX.
A glycoprotein that is important in the activation of CLASSICAL COMPLEMENT PATHWAY. C4 is cleaved by the activated COMPLEMENT C1S into COMPLEMENT C4A and COMPLEMENT C4B.
The sequential activation of serum COMPLEMENT PROTEINS to create the COMPLEMENT MEMBRANE ATTACK COMPLEX. Factors initiating complement activation include ANTIGEN-ANTIBODY COMPLEXES, microbial ANTIGENS, or cell surface POLYSACCHARIDES.
Serine proteases that cleave COMPLEMENT C3 into COMPLEMENT C3A and COMPLEMENT C3B, or cleave COMPLEMENT C5 into COMPLEMENT C5A and COMPLEMENT C5B. These include the different forms of C3/C5 convertases in the classical and the alternative pathways of COMPLEMENT ACTIVATION. Both cleavages take place at the C-terminal of an ARGININE residue.
The larger fragment generated from the cleavage of COMPLEMENT C3 by C3 CONVERTASE. It is a constituent of the ALTERNATIVE PATHWAY C3 CONVERTASE (C3bBb), and COMPLEMENT C5 CONVERTASES in both the classical (C4b2a3b) and the alternative (C3bBb3b) pathway. C3b participates in IMMUNE ADHERENCE REACTION and enhances PHAGOCYTOSIS. It can be inactivated (iC3b) or cleaved by various proteases to yield fragments such as COMPLEMENT C3C; COMPLEMENT C3D; C3e; C3f; and C3g.
A glycine-rich, heat-labile serum glycoprotein that contains a component of the C3 CONVERTASE ALTERNATE PATHWAY (C3bBb). Bb, a serine protease, is generated when factor B is cleaved by COMPLEMENT FACTOR D into Ba and Bb.
The smaller fragment formed when complement C4 is cleaved by COMPLEMENT C1S. It is an anaphylatoxin that causes symptoms of immediate hypersensitivity (HYPERSENSITIVITY, IMMEDIATE) but its activity is weaker than that of COMPLEMENT C3A or COMPLEMENT C5A.
C5 plays a central role in both the classical and the alternative pathway of COMPLEMENT ACTIVATION. C5 is cleaved by C5 CONVERTASE into COMPLEMENT C5A and COMPLEMENT C5B. The smaller fragment C5a is an ANAPHYLATOXIN and mediator of inflammatory process. The major fragment C5b binds to the membrane initiating the spontaneous assembly of the late complement components, C5-C9, into the MEMBRANE ATTACK COMPLEX.
A serine protease that is the complex of COMPLEMENT C3B and COMPLEMENT FACTOR BB. It cleaves multiple COMPLEMENT C3 into COMPLEMENT C3A (anaphylatoxin) and COMPLEMENT C3B in the ALTERNATIVE COMPLEMENT ACTIVATION PATHWAY.
The smaller fragment generated from the cleavage of complement C3 by C3 CONVERTASE. C3a, a 77-amino acid peptide, is a mediator of local inflammatory process. It induces smooth MUSCLE CONTRACTION, and HISTAMINE RELEASE from MAST CELLS and LEUKOCYTES. C3a is considered an anaphylatoxin along with COMPLEMENT C4A; COMPLEMENT C5A; and COMPLEMENT C5A, DES-ARGININE.
Serum glycoproteins participating in the host defense mechanism of COMPLEMENT ACTIVATION that creates the COMPLEMENT MEMBRANE ATTACK COMPLEX. Included are glycoproteins in the various pathways of complement activation (CLASSICAL COMPLEMENT PATHWAY; ALTERNATIVE COMPLEMENT PATHWAY; and LECTIN COMPLEMENT PATHWAY).
A subcomponent of complement C1, composed of six copies of three polypeptide chains (A, B, and C), each encoded by a separate gene (C1QA; C1QB; C1QC). This complex is arranged in nine subunits (six disulfide-linked dimers of A and B, and three disulfide-linked homodimers of C). C1q has binding sites for antibodies (the heavy chain of IMMUNOGLOBULIN G or IMMUNOGLOBULIN M). The interaction of C1q and immunoglobulin activates the two proenzymes COMPLEMENT C1R and COMPLEMENT C1S, thus initiating the cascade of COMPLEMENT ACTIVATION via the CLASSICAL COMPLEMENT PATHWAY.
Complement activation initiated by the binding of COMPLEMENT C1 to ANTIGEN-ANTIBODY COMPLEXES at the COMPLEMENT C1Q subunit. This leads to the sequential activation of COMPLEMENT C1R and COMPLEMENT C1S subunits. Activated C1s cleaves COMPLEMENT C4 and COMPLEMENT C2 forming the membrane-bound classical C3 CONVERTASE (C4B2A) and the subsequent C5 CONVERTASE (C4B2A3B) leading to cleavage of COMPLEMENT C5 and the assembly of COMPLEMENT MEMBRANE ATTACK COMPLEX.
The large fragment formed when COMPLEMENT C4 is cleaved by COMPLEMENT C1S. The membrane-bound C4b binds COMPLEMENT C2A, a SERINE PROTEASE, to form C4b2a (CLASSICAL PATHWAY C3 CONVERTASE) and subsequent C4b2a3b (CLASSICAL PATHWAY C5 CONVERTASE).
A serine endopeptidase that has specificity for cleavage at ARGININE. It cleaves a variety of prohormones including PRO-OPIOMELANOCORTIN, proluteinizing-hormone-releasing hormone, proenkephalins, prodynorphin, and PROINSULIN.
The minor fragment formed when C5 convertase cleaves C5 into C5a and COMPLEMENT C5B. C5a is a 74-amino-acid glycopeptide with a carboxy-terminal ARGININE that is crucial for its spasmogenic activity. Of all the complement-derived anaphylatoxins, C5a is the most potent in mediating immediate hypersensitivity (HYPERSENSITIVITY, IMMEDIATE), smooth MUSCLE CONTRACTION; HISTAMINE RELEASE; and migration of LEUKOCYTES to site of INFLAMMATION.
A component of the CLASSICAL COMPLEMENT PATHWAY. C2 is cleaved by activated COMPLEMENT C1S into COMPLEMENT C2B and COMPLEMENT C2A. C2a, the COOH-terminal fragment containing a SERINE PROTEASE, combines with COMPLEMENT C4B to form C4b2a (CLASSICAL PATHWAY C3 CONVERTASE) and subsequent C4b2a3b (CLASSICAL PATHWAY C5 CONVERTASE).
A CALCIUM-dependent endopeptidase that has specificity for cleavage at ARGININE that is near paired basic residues. It cleaves a variety of prohormones including PRO-OPIOMELANOCORTIN; PRORENIN; proenkephalins; prodynorphin; prosomatostatin; and PROINSULIN.
Enzymes that activate one or more COMPLEMENT PROTEINS in the complement system leading to the formation of the COMPLEMENT MEMBRANE ATTACK COMPLEX, an important response in host defense. They are enzymes in the various COMPLEMENT ACTIVATION pathways.
Serum proteins that negatively regulate the cascade process of COMPLEMENT ACTIVATION. Uncontrolled complement activation and resulting cell lysis is potentially dangerous for the host. The complement system is tightly regulated by inactivators that accelerate the decay of intermediates and certain cell surface receptors.
A 53-kDa protein that is a positive regulator of the alternate pathway of complement activation (COMPLEMENT ACTIVATION PATHWAY, ALTERNATIVE). It stabilizes the ALTERNATIVE PATHWAY C3 CONVERTASE (C3bBb) and protects it from rapid inactivation, thus facilitating the cascade of COMPLEMENT ACTIVATION and the formation of MEMBRANE ATTACK COMPLEX. Individuals with mutation in the PFC gene exhibit properdin deficiency and have a high susceptibility to infections.
A 105-kDa serum glycoprotein with significant homology to the other late complement components, C7-C9. It is a polypeptide chain cross-linked by 32 disulfide bonds. C6 is the next complement component to bind to the membrane-bound COMPLEMENT C5B in the assembly of MEMBRANE ATTACK COMPLEX. It is encoded by gene C6.
An important soluble regulator of the alternative pathway of complement activation (COMPLEMENT ACTIVATION PATHWAY, ALTERNATIVE). It is a 139-kDa glycoprotein expressed by the liver and secreted into the blood. It binds to COMPLEMENT C3B and makes iC3b (inactivated complement 3b) susceptible to cleavage by COMPLEMENT FACTOR I. Complement factor H also inhibits the association of C3b with COMPLEMENT FACTOR B to form the C3bB proenzyme, and promotes the dissociation of Bb from the C3bBb complex (COMPLEMENT C3 CONVERTASE, ALTERNATIVE PATHWAY).
A 302-amino-acid fragment in the alpha chain (672-1663) of C3b. It is generated when C3b is inactivated (iC3b) and its alpha chain is cleaved by COMPLEMENT FACTOR I into C3c, and C3dg (955-1303) in the presence COMPLEMENT FACTOR H. Serum proteases further degrade C3dg into C3d (1002-1303) and C3g (955-1001).
A serum protein which is important in the ALTERNATIVE COMPLEMENT ACTIVATION PATHWAY. This enzyme cleaves the COMPLEMENT C3B-bound COMPLEMENT FACTOR B to form C3bBb which is ALTERNATIVE PATHWAY C3 CONVERTASE.
A 206-amino-acid fragment in the alpha chain (672-1663) of C3b. It is generated when C3b is inactivated (iC3b) and its alpha chain is cleaved by COMPLEMENT FACTOR I into C3c (749-954), and C3dg (955-1303) in the presence COMPLEMENT FACTOR H.
A 63-kDa serum glycoprotein encoded by gene C9. Monomeric C9 (mC9) binds the C5b-8 complex to form C5b-9 which catalyzes the polymerization of C9 forming C5b-p9 (MEMBRANE ATTACK COMPLEX) and transmembrane channels leading to lysis of the target cell. Patients with C9 deficiency suffer from recurrent bacterial infections.
Molecules on the surface of some B-lymphocytes and macrophages, that recognize and combine with the C3b, C3d, C1q, and C4b components of complement.
A serine endopeptidase found primarily in the EXTRACELLULAR MATRIX. It has specificity for cleavage of a variety of substrates including PRORENIN, pro-membrane type-1 matrix metalloproteinase, and NEURAL CELL ADHESION MOLECULE L1.
Proteolytic enzymes that are involved in the conversion of protein precursors such as peptide prohormones into PEPTIDE HORMONES. Some are ENDOPEPTIDASES, some are EXOPEPTIDASES.
A product of COMPLEMENT ACTIVATION cascade, regardless of the pathways, that forms transmembrane channels causing disruption of the target CELL MEMBRANE and cell lysis. It is formed by the sequential assembly of terminal complement components (COMPLEMENT C5B; COMPLEMENT C6; COMPLEMENT C7; COMPLEMENT C8; and COMPLEMENT C9) into the target membrane. The resultant C5b-8-poly-C9 is the "membrane attack complex" or MAC.
A 77-kDa subcomponent of complement C1, encoded by gene C1S, is a SERINE PROTEASE existing as a proenzyme (homodimer) in the intact complement C1 complex. Upon the binding of COMPLEMENT C1Q to antibodies, the activated COMPLEMENT C1R cleaves C1s into two chains, A (heavy) and B (light, the serine protease), linked by disulfide bonds yielding the active C1s. The activated C1s, in turn, cleaves COMPLEMENT C2 and COMPLEMENT C4 to form C4b2a (CLASSICAL C3 CONVERTASE).
Endogenous proteins that inhibit or inactivate COMPLEMENT C3B. They include COMPLEMENT FACTOR H and COMPLEMENT FACTOR I (C3b/C4b inactivator). They cleave or promote the cleavage of C3b into inactive fragments, and thus are important in the down-regulation of COMPLEMENT ACTIVATION and its cytolytic sequence.
A 93-kDa serum glycoprotein encoded by C7 gene. It is a polypeptide chain with 28 disulfide bridges. In the formation of MEMBRANE ATTACK COMPLEX; C7 is the next component to bind the C5b-6 complex forming a trimolecular complex C5b-7 which is lipophilic, resembles an integral membrane protein, and serves as an anchor for the late complement components, C8 and C9.
The first complement component to act in the activation of CLASSICAL COMPLEMENT PATHWAY. It is a calcium-dependent trimolecular complex made up of three subcomponents: COMPLEMENT C1Q; COMPLEMENT C1R; and COMPLEMENT C1S at 1:2:2 ratios. When the intact C1 binds to at least two antibodies (involving C1q), C1r and C1s are sequentially activated, leading to subsequent steps in the cascade of COMPLEMENT ACTIVATION.
Molecular sites on or in some B-lymphocytes and macrophages that recognize and combine with COMPLEMENT C3B. The primary structure of these receptors reveal that they contain transmembrane and cytoplasmic domains, with their extracellular portion composed entirely of thirty short consensus repeats each having 60 to 70 amino acids.
An IgG autoantibody against the ALTERNATIVE PATHWAY C3 CONVERTASE, found in serum of patients with MESANGIOCAPILLARY GLOMERULONEPHRITIS. The binding of this autoantibody to C3bBb stabilizes the enzyme thus reduces the actions of C3b inactivators (COMPLEMENT FACTOR H; COMPLEMENT FACTOR I). This abnormally stabilized enzyme induces a continuous COMPLEMENT ACTIVATION and generation of C3b thereby promoting the assembly of MEMBRANE ATTACK COMPLEX and cytolysis.
A 80-kDa subcomponent of complement C1, existing as a SERINE PROTEASE proenzyme in the intact complement C1 complex. When COMPLEMENT C1Q is bound to antibodies, the changed tertiary structure causes autolytic activation of complement C1r which is cleaved into two chains, A (heavy) and B (light, the serine protease), connected by disulfide bonds. The activated C1r serine protease, in turn, activates COMPLEMENT C1S proenzyme by cleaving the Arg426-Ile427 bond. No fragment is released when either C1r or C1s is cleaved.
The larger fragment generated from the cleavage of C5 by C5 CONVERTASE that yields COMPLEMENT C5A and C5b (beta chain + alpha' chain, the residual alpha chain, bound by disulfide bond). C5b remains bound to the membrane and initiates the spontaneous assembly of the late complement components to form C5b-8-poly-C9, the MEMBRANE ATTACK COMPLEX.
A family of SERINE ENDOPEPTIDASES isolated from Bacillus subtilis. EC 3.4.21.-
A proprotein convertase with specificity for the proproteins of PROALBUMIN; COMPLEMENT 3C; and VON WILLEBRAND FACTOR. It has specificity for cleavage near paired ARGININE residues that are separated by two amino acids.
A 150-kDa serum glycoprotein composed of three subunits with each encoded by a different gene (C8A; C8B; and C8G). This heterotrimer contains a disulfide-linked C8alpha-C8gamma heterodimer and a noncovalently associated C8beta chain. C8 is the next component to bind the C5-7 complex forming C5b-8 that binds COMPLEMENT C9 and acts as a catalyst in the polymerization of C9.
GPI-linked membrane proteins broadly distributed among hematopoietic and non-hematopoietic cells. CD55 prevents the assembly of C3 CONVERTASE or accelerates the disassembly of preformed convertase, thus blocking the formation of the membrane attack complex.
The COOH-terminal fragment of COMPLEMENT 2, released by the action of activated COMPLEMENT C1S. It is a SERINE PROTEASE. C2a combines with COMPLEMENT C4B to form C4b2a (CLASSICAL PATHWAY C3 CONVERTASE) and subsequent C4b2a3b (CLASSICAL PATHWAY C5 CONVERTASE).
A process whereby multiple RNA transcripts are generated from a single gene. Alternative splicing involves the splicing together of other possible sets of EXONS during the processing of some, but not all, transcripts of the gene. Thus a particular exon may be connected to any one of several alternative exons to form a mature RNA. The alternative forms of mature MESSENGER RNA produce PROTEIN ISOFORMS in which one part of the isoforms is common while the other parts are different.
A plasma serine proteinase that cleaves the alpha-chains of C3b and C4b in the presence of the cofactors COMPLEMENT FACTOR H and C4-binding protein, respectively. It is a 66-kDa glycoprotein that converts C3b to inactivated C3b (iC3b) followed by the release of two fragments, C3c (150-kDa) and C3dg (41-kDa). It was formerly called KAF, C3bINF, or enzyme 3b inactivator.
Important enzymes in the ALTERNATIVE COMPLEMENT ACTIVATION PATHWAY. They cleave COMPLEMENT C3 and COMPLEMENT C5.
Compounds that negatively regulate the cascade process of COMPLEMENT ACTIVATION. Uncontrolled complement activation and resulting cell lysis is potentially dangerous for the host.
The destruction of ERYTHROCYTES by many different causal agents such as antibodies, bacteria, chemicals, temperature, and changes in tonicity.
A screening assay for circulating COMPLEMENT PROTEINS. Diluted SERUM samples are added to antibody-coated ERYTHROCYTES and the percentage of cell lysis is measured. The values are expressed by the so called CH50, in HEMOLYTIC COMPLEMENT units per milliliter, which is the dilution of serum required to lyse 50 percent of the erythrocytes in the assay.
A serine protease that cleaves multiple COMPLEMENT C5 into COMPLEMENT C5A (anaphylatoxin) and COMPLEMENT C5B in the ALTERNATIVE COMPLEMENT ACTIVATION PATHWAY. It is the complex of ALTERNATIVE PATHWAY C3 CONVERTASE (C3bBb) with an additional COMPLEMENT C3B, or C3bBb3b.
Molecular sites on or in B-lymphocytes, follicular dendritic cells, lymphoid cells, and epithelial cells that recognize and combine with COMPLEMENT C3D. Human complement receptor 2 (CR2) serves as a receptor for both C3dg and the gp350/220 glycoprotein of HERPESVIRUS 4, HUMAN, and binds the monoclonal antibody OKB7, which blocks binding of both ligands to the receptor.
Venoms from snakes of the genus Naja (family Elapidae). They contain many specific proteins that have cytotoxic, hemolytic, neurotoxic, and other properties. Like other elapid venoms, they are rich in enzymes. They include cobramines and cobralysins.
A G-protein-coupled receptor that signals an increase in intracellular calcium in response to the potent ANAPHYLATOXIN peptide COMPLEMENT C5A.
A serum protein that regulates the CLASSICAL COMPLEMENT ACTIVATION PATHWAY. It binds as a cofactor to COMPLEMENT FACTOR I which then hydrolyzes the COMPLEMENT C4B in the CLASSICAL PATHWAY C3 CONVERTASE (C4bC2a).
Serum peptides derived from certain cleaved COMPLEMENT PROTEINS during COMPLEMENT ACTIVATION. They induce smooth MUSCLE CONTRACTION; mast cell HISTAMINE RELEASE; PLATELET AGGREGATION; and act as mediators of the local inflammatory process. The order of anaphylatoxin activity from the strongest to the weakest is C5a, C3a, C4a, and C5a des-arginine.
The N-terminal fragment of COMPLEMENT 2, released by the action of activated COMPLEMENT C1S.
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.
Serum proteins that inhibit, antagonize, or inactivate COMPLEMENT C1 or its subunits.
A serine protease that cleaves multiple COMPLEMENT 3 into COMPLEMENT 3A (anaphylatoxin) and COMPLEMENT 3B in the CLASSICAL COMPLEMENT ACTIVATION PATHWAY. It is a complex of COMPLEMENT 4B and COMPLEMENT 2A (C4b2a).
Any member of the group of ENDOPEPTIDASES containing at the active site a serine residue involved in catalysis.
An acidic protein found in the NEUROENDOCRINE SYSTEM that functions as a molecular chaperone for PROPROTEIN CONVERTASE 2.
Proteins that bind to particles and cells to increase susceptibility to PHAGOCYTOSIS, especially ANTIBODIES bound to EPITOPES that attach to FC RECEPTORS. COMPLEMENT C3B may also participate.
The 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.
The complex formed by the binding of antigen and antibody molecules. The deposition of large antigen-antibody complexes leading to tissue damage causes IMMUNE COMPLEX DISEASES.
A serine protease that cleaves multiple COMPLEMENT 5 into COMPLEMENT 5A (anaphylatoxin) and COMPLEMENT 5B in the CLASSICAL COMPLEMENT ACTIVATION PATHWAY. It is a complex of CLASSICAL PATHWAY C3 CONVERTASE (C4b2a) with an additional COMPLEMENT C3B, or C4b2a3b.
Zymosan is a polysaccharide derived from the cell walls of Saccharomyces cerevisiae, commonly used in research as an immunostimulant to induce inflammation and study phagocytosis, complement activation, and oxidative burst in neutrophils and macrophages.
Serologic tests based on inactivation of complement by the antigen-antibody complex (stage 1). Binding of free complement can be visualized by addition of a second antigen-antibody system such as red cells and appropriate red cell antibody (hemolysin) requiring complement for its completion (stage 2). Failure of the red cells to lyse indicates that a specific antigen-antibody reaction has taken place in stage 1. If red cells lyse, free complement is present indicating no antigen-antibody reaction occurred in stage 1.
Small glycoproteins found on both hematopoietic and non-hematopoietic cells. CD59 restricts the cytolytic activity of homologous complement by binding to C8 and C9 and blocking the assembly of the membrane attack complex. (From Barclay et al., The Leukocyte Antigen FactsBook, 1993, p234)
Complement activation triggered by the interaction of microbial POLYSACCHARIDES with serum MANNOSE-BINDING LECTIN resulting in the activation of MANNOSE-BINDING PROTEIN-ASSOCIATED SERINE PROTEASES. As in the classical pathway, MASPs cleave COMPLEMENT C4 and COMPLEMENT C2 to form C3 CONVERTASE (C4B2A) and the subsequent C5 CONVERTASE (C4B2A3B) leading to cleavage of COMPLEMENT C5 and assembly of COMPLEMENT MEMBRANE ATTACK COMPLEX.
A ubiquitously expressed complement receptor that binds COMPLEMENT C3B and COMPLEMENT C4B and serves as a cofactor for their inactivation. CD46 also interacts with a wide variety of pathogens and mediates immune response.
A ZINC-containing exopeptidase primarily found in SECRETORY VESICLES of endocrine and neuroendocrine cells. It catalyzes the cleavage of C-terminal ARGININE or LYSINE residues from polypeptides and is active in processing precursors of PEPTIDE HORMONES and other bioactive peptides.
The major immunoglobulin isotype class in normal human serum. There are several isotype subclasses of IgG, for example, IgG1, IgG2A, and IgG2B.
The sequence of PURINES and PYRIMIDINES in nucleic acids and polynucleotides. It is also called nucleotide sequence.
The process in which substances, either endogenous or exogenous, bind to proteins, peptides, enzymes, protein precursors, or allied compounds. Specific protein-binding measures are often used as assays in diagnostic assessments.
An endogenous 105-kDa plasma glycoprotein produced primarily by the LIVER and MONOCYTES. It inhibits a broad spectrum of proteases, including the COMPLEMENT C1R and the COMPLEMENT C1S proteases of the CLASSICAL COMPLEMENT PATHWAY, and the MANNOSE-BINDING PROTEIN-ASSOCIATED SERINE PROTEASES. C1-INH-deficient individuals suffer from HEREDITARY ANGIOEDEMA TYPES I AND II.
Chronic glomerulonephritis characterized histologically by proliferation of MESANGIAL CELLS, increase in the MESANGIAL EXTRACELLULAR MATRIX, and a thickening of the glomerular capillary walls. This may appear as a primary disorder or secondary to other diseases including infections and autoimmune disease SYSTEMIC LUPUS ERYTHEMATOSUS. Various subtypes are classified by their abnormal ultrastructures and immune deposits. Hypocomplementemia is a characteristic feature of all types of MPGN.
Red blood cells. Mature erythrocytes are non-nucleated, biconcave disks containing HEMOGLOBIN whose function is to transport OXYGEN.
The engulfing and degradation of microorganisms; other cells that are dead, dying, or pathogenic; and foreign particles by phagocytic cells (PHAGOCYTES).
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.
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.
Any detectable and heritable change in the genetic material that causes a change in the GENOTYPE and which is transmitted to daughter cells and to succeeding generations.
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.
Established cell cultures that have the potential to propagate indefinitely.
Important enzymes in the CLASSICAL COMPLEMENT ACTIVATION PATHWAY. They cleave COMPLEMENT C3 and COMPLEMENT C5.
The rate dynamics in chemical or physical systems.
Physiologically inactive substances that can be converted to active enzymes.
Proteins prepared by recombinant DNA technology.
Proteins that are present in blood serum, including SERUM ALBUMIN; BLOOD COAGULATION FACTORS; and many other types of proteins.
The natural bactericidal property of BLOOD due to normally occurring antibacterial substances such as beta lysin, leukin, etc. This activity needs to be distinguished from the bactericidal activity contained in a patient's serum as a result of antimicrobial therapy, which is measured by a SERUM BACTERICIDAL TEST.
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.
The parts of a macromolecule that directly participate in its specific combination with another molecule.
Protein precursors, also known as proproteins or prohormones, are inactive forms of proteins that undergo post-translational modification, such as cleavage, to produce the active functional protein or peptide hormone.
The level of protein structure in which combinations of secondary protein structures (alpha helices, beta sheets, loop regions, and motifs) pack together to form folded shapes called domains. Disulfide bridges between cysteines in two different parts of the polypeptide chain along with other interactions between the chains play a role in the formation and stabilization of tertiary structure. Small proteins usually consist of only one domain but larger proteins may contain a number of domains connected by segments of polypeptide chain which lack regular secondary structure.
Inflammation of the renal glomeruli (KIDNEY GLOMERULUS) that can be classified by the type of glomerular injuries including antibody deposition, complement activation, cellular proliferation, and glomerulosclerosis. These structural and functional abnormalities usually lead to HEMATURIA; PROTEINURIA; HYPERTENSION; and RENAL INSUFFICIENCY.
The species Oryctolagus cuniculus, in the family Leporidae, order LAGOMORPHA. Rabbits are born in burrows, furless, and with eyes and ears closed. In contrast with HARES, rabbits have 22 chromosome pairs.
Any of various enzymatically catalyzed post-translational modifications of PEPTIDES or PROTEINS in the cell of origin. These modifications include carboxylation; HYDROXYLATION; ACETYLATION; PHOSPHORYLATION; METHYLATION; GLYCOSYLATION; ubiquitination; oxidation; proteolysis; and crosslinking and result in changes in molecular weight and electrophoretic motility.
Serum serine proteases which participate in COMPLEMENT ACTIVATION. They are activated when complexed with the MANNOSE-BINDING LECTIN, therefore also known as Mannose-binding protein-Associated Serine Proteases (MASPs). They cleave COMPLEMENT C4 and COMPLEMENT C2 to form C4b2a, the CLASSICAL PATHWAY C3 CONVERTASE.
Partial proteins formed by partial hydrolysis of complete proteins or generated through PROTEIN ENGINEERING techniques.
An adrenal microsomal cytochrome P450 enzyme that catalyzes the 21-hydroxylation of steroids in the presence of molecular oxygen and NADPH-FERRIHEMOPROTEIN REDUCTASE. This enzyme, encoded by CYP21 gene, converts progesterones to precursors of adrenal steroid hormones (CORTICOSTERONE; HYDROCORTISONE). Defects in CYP21 cause congenital adrenal hyperplasia (ADRENAL HYPERPLASIA, CONGENITAL).
A specific mannose-binding member of the collectin family of lectins. It binds to carbohydrate groups on invading pathogens and plays a key role in the MANNOSE-BINDING LECTIN COMPLEMENT PATHWAY.
Granular leukocytes having a nucleus with three to five lobes connected by slender threads of chromatin, and cytoplasm containing fine inconspicuous granules and stainable by neutral dyes.
A chronic, relapsing, inflammatory, and often febrile multisystemic disorder of connective tissue, characterized principally by involvement of the skin, joints, kidneys, and serosal membranes. It is of unknown etiology, but is thought to represent a failure of the regulatory mechanisms of the autoimmune system. The disease is marked by a wide range of system dysfunctions, an elevated erythrocyte sedimentation rate, and the formation of LE cells in the blood or bone marrow.
A condition characterized by the recurrence of HEMOGLOBINURIA caused by intravascular HEMOLYSIS. In cases occurring upon cold exposure (paroxysmal cold hemoglobinuria), usually after infections, there is a circulating antibody which is also a cold hemolysin. In cases occurring during or after sleep (paroxysmal nocturnal hemoglobinuria), the clonal hematopoietic stem cells exhibit a global deficiency of cell membrane proteins.
Antibodies produced by a single clone of cells.
Esterases are hydrolase enzymes that catalyze the hydrolysis of ester bonds, converting esters into alcohols and acids, playing crucial roles in various biological processes including metabolism and detoxification.
The degree of similarity between sequences of amino acids. This information is useful for the analyzing genetic relatedness of proteins and species.
Electrophoresis in which a polyacrylamide gel is used as the diffusion medium.
The insertion of recombinant DNA molecules from prokaryotic and/or eukaryotic sources into a replicating vehicle, such as a plasmid or virus vector, and the introduction of the resultant hybrid molecules into recipient cells without altering the viability of those cells.
Identification of proteins or peptides that have been electrophoretically separated by blot transferring from the electrophoresis gel to strips of nitrocellulose paper, followed by labeling with antibody probes.
The clear portion of BLOOD that is left after BLOOD COAGULATION to remove BLOOD CELLS and clotting proteins.
A syndrome that is associated with microvascular diseases of the KIDNEY, such as RENAL CORTICAL NECROSIS. It is characterized by hemolytic anemia (ANEMIA, HEMOLYTIC); THROMBOCYTOPENIA; and ACUTE RENAL FAILURE.
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.
Elements of limited time intervals, contributing to particular results or situations.
An adhesion-promoting leukocyte surface membrane heterodimer. The alpha subunit consists of the CD11b ANTIGEN and the beta subunit the CD18 ANTIGEN. The antigen, which is an integrin, functions both as a receptor for complement 3 and in cell-cell and cell-substrate adhesive interactions.
A technique that combines protein electrophoresis and double immunodiffusion. In this procedure proteins are first separated by gel electrophoresis (usually agarose), then made visible by immunodiffusion of specific antibodies. A distinct elliptical precipitin arc results for each protein detectable by the antisera.
A chelating agent that sequesters a variety of polyvalent cations such as CALCIUM. It is used in pharmaceutical manufacturing and as a food additive.
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.
A cluster of convoluted capillaries beginning at each nephric tubule in the kidney and held together by connective tissue.
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.
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 sub-subclass of endopeptidases that depend on an ASPARTIC ACID residue for their activity.
Hormones secreted by the PITUITARY GLAND including those from the anterior lobe (adenohypophysis), the posterior lobe (neurohypophysis), and the ill-defined intermediate lobe. Structurally, they include small peptides, proteins, and glycoproteins. They are under the regulation of neural signals (NEUROTRANSMITTERS) or neuroendocrine signals (HYPOTHALAMIC HORMONES) from the hypothalamus as well as feedback from their targets such as ADRENAL CORTEX HORMONES; ANDROGENS; ESTROGENS.
A class of C-type lectins that target the carbohydrate structures found on invading pathogens. Binding of collectins to microorganisms results in their agglutination and enhanced clearance. Collectins form trimers that may assemble into larger oligomers. Each collectin polypeptide chain consists of four regions: a relatively short N-terminal region, a collagen-like region, an alpha-helical coiled-coil region, and carbohydrate-binding region.
The uptake of naked or purified DNA by CELLS, usually meaning the process as it occurs in eukaryotic cells. It is analogous to bacterial transformation (TRANSFORMATION, BACTERIAL) and both are routinely employed in GENE TRANSFER TECHNIQUES.
The common precursor polypeptide of pancreatic GLUCAGON and intestinal GLUCAGON-LIKE PEPTIDES. Proglucagon is the 158-amino acid segment of preproglucagon without the N-terminal signal sequence. Proglucagon is expressed in the PANCREAS; INTESTINES; and the CENTRAL NERVOUS SYSTEM. Posttranslational processing of proglucagon is tissue-specific yielding numerous bioactive peptides.
A biosensing technique in which biomolecules capable of binding to specific analytes or ligands are first immobilized on one side of a metallic film. Light is then focused on the opposite side of the film to excite the surface plasmons, that is, the oscillations of free electrons propagating along the film's surface. The refractive index of light reflecting off this surface is measured. When the immobilized biomolecules are bound by their ligands, an alteration in surface plasmons on the opposite side of the film is created which is directly proportional to the change in bound, or adsorbed, mass. Binding is measured by changes in the refractive index. The technique is used to study biomolecular interactions, such as antigen-antibody binding.
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.
The parts of a transcript of a split GENE remaining after the INTRONS are removed. They are spliced together to become a MESSENGER RNA or other functional RNA.
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.
Immunoglobulin molecules having a specific amino acid sequence by virtue of which they interact only with the ANTIGEN (or a very similar shape) that induced their synthesis in cells of the lymphoid series (especially PLASMA CELLS).
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.
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.
In vitro method for producing large amounts of specific DNA or RNA fragments of defined length and sequence from small amounts of short oligonucleotide flanking sequences (primers). The essential steps include thermal denaturation of the double-stranded target molecules, annealing of the primers to their complementary sequences, and extension of the annealed primers by enzymatic synthesis with DNA polymerase. The reaction is efficient, specific, and extremely sensitive. Uses for the reaction include disease diagnosis, detection of difficult-to-isolate pathogens, mutation analysis, genetic testing, DNA sequencing, and analyzing evolutionary relationships.
Conjugated protein-carbohydrate compounds including mucins, mucoid, and amyloid glycoproteins.
A common name used for the genus Cavia. The most common species is Cavia porcellus which is the domesticated guinea pig used for pets and biomedical research.
A subfamily in the family MURIDAE, comprising the hamsters. Four of the more common genera are Cricetus, CRICETULUS; MESOCRICETUS; and PHODOPUS.
Short sequences (generally about 10 base pairs) of DNA that are complementary to sequences of messenger RNA and allow reverse transcriptases to start copying the adjacent sequences of mRNA. Primers are used extensively in genetic and molecular biology techniques.
A 30-kDa protein synthesized primarily in the ANTERIOR PITUITARY GLAND and the HYPOTHALAMUS. It is also found in the skin and other peripheral tissues. Depending on species and tissues, POMC is cleaved by PROHORMONE CONVERTASES yielding various active peptides including ACTH; BETA-LIPOTROPIN; ENDORPHINS; MELANOCYTE-STIMULATING HORMONES; and others (GAMMA-LPH; CORTICOTROPIN-LIKE INTERMEDIATE LOBE PEPTIDE; N-terminal peptide of POMC or NPP).
Transport proteins that carry specific substances in the blood or across cell membranes.
The sum of the weight of all the atoms in a molecule.
A specific immune response elicited by a specific dose of an immunologically active substance or cell in an organism, tissue, or cell.
CELL LINE derived from the ovary of the Chinese hamster, Cricetulus griseus (CRICETULUS). The species is a favorite for cytogenetic studies because of its small chromosome number. The cell line has provided model systems for the study of genetic alterations in cultured mammalian cells.
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.)
A deoxyribonucleotide polymer that is the primary genetic material of all cells. Eukaryotic and prokaryotic organisms normally contain DNA in a double-stranded state, yet several important biological processes transiently involve single-stranded regions. DNA, which consists of a polysugar-phosphate backbone possessing projections of purines (adenine and guanine) and pyrimidines (thymine and cytosine), forms a double helix that is held together by hydrogen bonds between these purines and pyrimidines (adenine to thymine and guanine to cytosine).
The phenotypic manifestation of a gene or genes by the processes of GENETIC TRANSCRIPTION and GENETIC TRANSLATION.
The outward appearance of the individual. It is the product of interactions between genes, and between the GENOTYPE and the environment.
Proteins found in any species of bacterium.
A test used to determine whether or not complementation (compensation in the form of dominance) will occur in a cell with a given mutant phenotype when another mutant genome, encoding the same mutant phenotype, is introduced into that cell.
Single-stranded complementary DNA synthesized from an RNA template by the action of RNA-dependent DNA polymerase. cDNA (i.e., complementary DNA, not circular DNA, not C-DNA) is used in a variety of molecular cloning experiments as well as serving as a specific hybridization probe.
Glycoproteins found on the membrane or surface of cells.
Polysaccharides consisting of mannose units.
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.
Theoretical representations that simulate the behavior or activity of biological processes or diseases. For disease models in living animals, DISEASE MODELS, ANIMAL is available. Biological models include the use of mathematical equations, computers, and other electronic equipment.
A characteristic feature of enzyme activity in relation to the kind of substrate on which the enzyme or catalytic molecule reacts.
Immunoelectrophoresis in which a second electrophoretic transport is performed on the initially separated antigen fragments into an antibody-containing medium in a direction perpendicular to the first electrophoresis.
A derivative of complement C5a, generated when the carboxy-terminal ARGININE is removed by CARBOXYPEPTIDASE B present in normal human serum. C5a des-Arg shows complete loss of spasmogenic activity though it retains some chemotactic ability (CHEMOATTRACTANTS).
A chelating agent relatively more specific for calcium and less toxic than EDETIC ACID.
Inflammation of any part of the KIDNEY.
Models used experimentally or theoretically to study molecular shape, electronic properties, or interactions; includes analogous molecules, computer-generated graphics, and mechanical structures.
Histochemical localization of immunoreactive substances using labeled antibodies as reagents.
Plasma glycoprotein clotted by thrombin, composed of a dimer of three non-identical pairs of polypeptide chains (alpha, beta, gamma) held together by disulfide bonds. Fibrinogen clotting is a sol-gel change involving complex molecular arrangements: whereas fibrinogen is cleaved by thrombin to form polypeptides A and B, the proteolytic action of other enzymes yields different fibrinogen degradation products.
The restriction of a characteristic behavior, anatomical structure or physical system, such as immune response; metabolic response, or gene or gene variant to the members of one species. It refers to that property which differentiates one species from another but it is also used for phylogenetic levels higher or lower than the species.
Lipid-containing polysaccharides which are endotoxins and important group-specific antigens. They are often derived from the cell wall of gram-negative bacteria and induce immunoglobulin secretion. The lipopolysaccharide molecule consists of three parts: LIPID A, core polysaccharide, and O-specific chains (O ANTIGENS). When derived from Escherichia coli, lipopolysaccharides serve as polyclonal B-cell mitogens commonly used in laboratory immunology. (From Dorland, 28th ed)
A large lobed glandular organ in the abdomen of vertebrates that is responsible for detoxification, metabolism, synthesis and storage of various substances.
Serum proteins with an electrophoretic mobility that falls between ALPHA-GLOBULINS and GAMMA-GLOBULINS.
A variation of the PCR technique in which cDNA is made from RNA via reverse transcription. The resultant cDNA is then amplified using standard PCR protocols.
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.
The lipid- and protein-containing, selectively permeable membrane that surrounds the cytoplasm in prokaryotic and eukaryotic cells.
Antibodies that react with self-antigens (AUTOANTIGENS) of the organism that produced them.
Body organ that filters blood for the secretion of URINE and that regulates ion concentrations.
ENDOPEPTIDASES which use a metal such as ZINC in the catalytic mechanism.
The biosynthesis of RNA carried out on a template of DNA. The biosynthesis of DNA from an RNA template is called REVERSE TRANSCRIPTION.
The arrangement of two or more amino acid or base sequences from an organism or organisms in such a way as to align areas of the sequences sharing common properties. The degree of relatedness or homology between the sequences is predicted computationally or statistically based on weights assigned to the elements aligned between the sequences. This in turn can serve as a potential indicator of the genetic relatedness between the organisms.
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.
The class of all enzymes catalyzing oxidoreduction reactions. The substrate that is oxidized is regarded as a hydrogen donor. The systematic name is based on donor:acceptor oxidoreductase. The recommended name will be dehydrogenase, wherever this is possible; as an alternative, reductase can be used. Oxidase is only used in cases where O2 is the acceptor. (Enzyme Nomenclature, 1992, p9)
A species of gram-negative, facultatively anaerobic, rod-shaped bacteria (GRAM-NEGATIVE FACULTATIVELY ANAEROBIC RODS) commonly found in the lower part of the intestine of warm-blooded animals. It is usually nonpathogenic, but some strains are known to produce DIARRHEA and pyogenic infections. Pathogenic strains (virotypes) are classified by their specific pathogenic mechanisms such as toxins (ENTEROTOXIGENIC ESCHERICHIA COLI), etc.
Detection of RNA that has been electrophoretically separated and immobilized by blotting on nitrocellulose or other type of paper or nylon membrane followed by hybridization with labeled NUCLEIC ACID PROBES.
A component of NF-kappa B transcription factor. It is proteolytically processed from NF-kappa B p100 precursor protein and is important for maturation of B-LYMPHOCYTES and adaptive HUMORAL IMMUNITY.
The relationship between the chemical structure of a compound and its biological or pharmacological activity. Compounds are often classed together because they have structural characteristics in common including shape, size, stereochemical arrangement, and distribution of functional groups.
Variant forms of the same gene, occupying the same locus on homologous CHROMOSOMES, and governing the variants in production of the same gene product.
A subclass of PEPTIDE HYDROLASES that catalyze the internal cleavage of PEPTIDES or PROTEINS.
The process of cleaving a chemical compound by the addition of a molecule of water.
A pathological process characterized by injury or destruction of tissues caused by a variety of cytologic and chemical reactions. It is usually manifested by typical signs of pain, heat, redness, swelling, and loss of function.
Enzymes that act at a free C-terminus of a polypeptide to liberate a single amino acid residue.
Polysaccharide isolated from the edible mushroom LENTINULA EDODES. The exact composition is unknown.
Recombinant proteins produced by the GENETIC TRANSLATION of fused genes formed by the combination of NUCLEIC ACID REGULATORY SEQUENCES of one or more genes with the protein coding sequences of one or more genes.
A 69-amino acid peptide derived from the N-terminal of PROGLUCAGON. It is mainly produced by the INTESTINAL L CELLS. Further processing of glicentin yield a 30-amino acid N-terminal peptide (glicentin-related polypeptide) and a 37-amino acid peptide OXYNTOMODULIN. Both glicentin and oxyntomodulin can reduce digestive secretions and delay gastric emptying.
A genus of trematode flukes belonging to the family Schistosomatidae. There are over a dozen species. These parasites are found in man and other mammals. Snails are the intermediate hosts.
Procedures, such as TISSUE CULTURE TECHNIQUES; mathematical models; etc., when used or advocated for use in place of the use of animals in research or diagnostic laboratories.
A serine endopeptidase isolated from Bacillus subtilis. It hydrolyzes proteins with broad specificity for peptide bonds, and a preference for a large uncharged residue in P1. It also hydrolyzes peptide amides. (From Enzyme Nomenclature, 1992) EC 3.4.21.62.
The characteristic 3-dimensional shape of a protein, including the secondary, supersecondary (motifs), tertiary (domains) and quaternary structure of the peptide chain. PROTEIN STRUCTURE, QUATERNARY describes the conformation assumed by multimeric proteins (aggregates of more than one polypeptide chain).
The relationship between the dose of an administered drug and the response of the organism to the drug.
A group of naturally occurring N-and O-acyl derivatives of the deoxyamino sugar neuraminic acid. They are ubiquitously distributed in many tissues.
Degenerative changes in the RETINA usually of older adults which results in a loss of vision in the center of the visual field (the MACULA LUTEA) because of damage to the retina. It occurs in dry and wet forms.
A metallic element that has the atomic symbol Mg, atomic number 12, and atomic weight 24.31. It is important for the activity of many enzymes, especially those involved in OXIDATIVE PHOSPHORYLATION.
A species of motile, free-living, gram-negative bacteria that occur in the soil. They are aerobic or microaerophilic and are sometimes capable of nitrogen fixation.
Thickening of the walls of small ARTERIES or ARTERIOLES due to cell proliferation or HYALINE deposition.
A highly acidic mucopolysaccharide formed of equal parts of sulfated D-glucosamine and D-glucuronic acid with sulfaminic bridges. The molecular weight ranges from six to twenty thousand. Heparin occurs in and is obtained from liver, lung, mast cells, etc., of vertebrates. Its function is unknown, but it is used to prevent blood clotting in vivo and vitro, in the form of many different salts.
A gram-positive organism found in the upper respiratory tract, inflammatory exudates, and various body fluids of normal and/or diseased humans and, rarely, domestic animals.
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.

Structural and functional analysis of a C3b-specific antibody that selectively inhibits the alternative pathway of complement. (1/1)

 (+info)

Complement C3 is a protein that plays a central role in the complement system, which is a part of the immune system that helps to clear pathogens and damaged cells from the body. Complement C3 can be activated through three different pathways: the classical pathway, the lectin pathway, and the alternative pathway. Once activated, it breaks down into two fragments, C3a and C3b.

C3a is an anaphylatoxin that helps to recruit immune cells to the site of infection or injury, while C3b plays a role in opsonization, which is the process of coating pathogens or damaged cells with proteins to make them more recognizable to the immune system. Additionally, C3b can also activate the membrane attack complex (MAC), which forms a pore in the membrane of target cells leading to their lysis or destruction.

In summary, Complement C3 is an important protein in the complement system that helps to identify and eliminate pathogens and damaged cells from the body through various mechanisms.

The alternative complement pathway is one of the three initiating pathways of the complement system, which is a part of the innate immune system that helps to clear pathogens and damaged cells from the body. The other two pathways are the classical and lectin pathways.

The alternative pathway is continuously activated at a low level, even in the absence of infection or injury, through the spontaneous cleavage of complement component C3 into C3a and C3b by the protease factor D in the presence of magnesium ions. The generated C3b can then bind covalently to nearby surfaces, including pathogens and host cells.

On self-surfaces, regulatory proteins like decay-accelerating factor (DAF) or complement receptor 1 (CR1) help to prevent the formation of the alternative pathway convertase and thus further activation of the complement system. However, on foreign surfaces, the C3b can recruit more complement components, forming a complex called the alternative pathway convertase (C3bBb), which cleaves additional C3 molecules into C3a and C3b.

The generated C3b can then bind to the surface and participate in the formation of the membrane attack complex (MAC), leading to the lysis of the target cell. The alternative pathway plays a crucial role in the defense against gram-negative bacteria, fungi, and parasites, as well as in the clearance of immune complexes and apoptotic cells. Dysregulation of the alternative complement pathway has been implicated in several diseases, including autoimmune disorders and atypical hemolytic uremic syndrome (aHUS).

Complement C4 is a protein that plays a crucial role in the complement system, which is a part of the immune system that helps to clear pathogens and damaged cells from the body. Complement C4 is involved in the early stages of the complement activation cascade, where it helps to identify and tag foreign or abnormal cells for destruction by other components of the immune system.

Specifically, Complement C4 can be cleaved into two smaller proteins, C4a and C4b, during the complement activation process. C4b then binds to the surface of the target cell and helps to initiate the formation of the membrane attack complex (MAC), which creates a pore in the cell membrane and leads to lysis or destruction of the target cell.

Deficiencies or mutations in the Complement C4 gene can lead to various immune disorders, including certain forms of autoimmune diseases and susceptibility to certain infections.

Complement activation is the process by which the complement system, a part of the immune system, is activated to help eliminate pathogens and damaged cells from the body. The complement system consists of a group of proteins that work together to recognize and destroy foreign substances.

Activation of the complement system can occur through three different pathways: the classical pathway, the lectin pathway, and the alternative pathway. Each pathway involves a series of proteolytic reactions that ultimately result in the formation of the membrane attack complex (MAC), which creates a pore in the membrane of the target cell, leading to its lysis and removal.

The classical pathway is typically activated by the binding of antibodies to antigens on the surface of a pathogen or damaged cell. The lectin pathway is activated by the recognition of specific carbohydrate structures on the surface of microorganisms. The alternative pathway can be spontaneously activated and serves as an amplification loop for both the classical and lectin pathways.

Complement activation plays a crucial role in the immune response, but uncontrolled or excessive activation can also lead to tissue damage and inflammation. Dysregulation of complement activation has been implicated in various diseases, including autoimmune disorders, inflammatory conditions, and neurodegenerative diseases.

Complement C3-C5 convertases are proteins that play a crucial role in the activation of the complement system, which is a part of the immune system. The complement system helps to eliminate pathogens and damaged cells from the body by marking them for destruction and attracting immune cells to the site of infection or injury.

The C3-C5 convertases are formed during the activation of the complement component 3 (C3) protein, which is a central player in the complement system. The formation of the C3-C5 convertase involves two main steps:

1. C3 convertase formation: In this step, a complex of proteins called the C3 convertase is formed by the cleavage of C3 into C3a and C3b fragments. This complex can then cleave additional C3 molecules into C3a and C3b fragments, amplifying the complement response.
2. C5 convertase formation: In this step, the C3b fragment from the C3 convertase binds to another protein called C4b2a, forming a new complex called the C5 convertase. The C5 convertase can then cleave the C5 protein into C5a and C5b fragments.

The C5b fragment goes on to form the membrane attack complex (MAC), which creates a pore in the membrane of the target cell, leading to its lysis or destruction. The C3a and C5a fragments are small proteins called anaphylatoxins that can cause inflammation and attract immune cells to the site of infection or injury.

Overall, the formation of Complement C3-C5 convertases is a critical step in the activation of the complement system and plays a key role in the body's defense against pathogens and damaged cells.

Complement C3b is a protein fragment that plays a crucial role in the complement system, which is a part of the immune system that helps to clear pathogens and damaged cells from the body. C3b is generated during the activation of the complement system, particularly via the classical, lectin, and alternative pathways.

Once formed, C3b can bind covalently to the surface of microbes or other target particles, marking them for destruction by other components of the immune system. Additionally, C3b can interact with other proteins in the complement system to generate the membrane attack complex (MAC), which forms pores in the membranes of targeted cells, leading to their lysis and removal.

In summary, Complement C3b is a vital protein fragment involved in the recognition, tagging, and elimination of pathogens and damaged cells during the immune response.

Complement Factor B is a protein that plays a crucial role in the complement system, which is a part of the immune system that helps to eliminate pathogens and damaged cells from the body. Specifically, Factor B is a component of the alternative pathway of the complement system, which provides a rapid and amplified response to microbial surfaces.

Factor B is cleaved by another protease called Factor D into two fragments, Ba and Bb. The formation of the C3 convertase (C3bBb) is essential for the activation of the alternative pathway. This complex can cleave and activate more C3 molecules, leading to a cascade of reactions that result in the formation of the membrane attack complex (MAC), which forms pores in the membranes of target cells, causing their lysis and elimination.

Deficiencies or mutations in Complement Factor B can lead to various complement-mediated diseases, such as atypical hemolytic uremic syndrome (aHUS) and age-related macular degeneration (AMD).

Complement C4a is a protein fragment or cleavage product generated during the activation of the complement system, which is a part of the immune system. The complement system helps to eliminate pathogens and damaged cells by marking them for destruction and direct lysis. Complement component 4 (C4) is one of the key proteins in this cascade, and it gets cleaved into C4a and C4b during the activation process.

C4a is a small anaphylatoxin with a molecular weight of approximately 9 kDa. It has chemotactic properties, meaning it can attract immune cells like neutrophils to the site of complement activation. Additionally, C4a can induce histamine release from mast cells and basophils, contributing to local inflammation. However, its precise physiological role in the immune response is not entirely clear, and dysregulation of C4a production has been implicated in several pathological conditions, such as autoimmune diseases and allergies.

Complement C5 is a protein that plays a crucial role in the complement system, which is a part of the immune system that helps to eliminate pathogens and damaged cells from the body. The complement system is a complex series of biochemical reactions that help to identify and destroy foreign substances, such as bacteria and viruses.

Complement C5 is one of several proteins in the complement system that are activated in a cascading manner in response to an activating event, such as the binding of an antibody to a pathogen. Once activated, Complement C5 can be cleaved into two smaller proteins, C5a and C5b.

C5a is a powerful anaphylatoxin, which means it can cause the release of histamine from mast cells and basophils, leading to inflammation and increased vascular permeability. It also acts as a chemoattractant, drawing immune cells to the site of infection or injury.

C5b, on the other hand, plays a role in the formation of the membrane attack complex (MAC), which is a protein structure that can punch holes in the membranes of pathogens, leading to their lysis and destruction.

Overall, Complement C5 is an important component of the immune system's response to infection and injury, helping to eliminate pathogens and damaged cells from the body.

Complement C3 Convertase, Alternative Pathway is a complex enzyme composed of the proteins C3b and Bb. It plays a crucial role in the alternative pathway of the complement system, which is a part of the innate immune system that helps to defend the body against invading pathogens.

The alternative pathway is continuously activated at a low level, and C3 Convertase is responsible for amplifying this activation. It does so by cleaving the complement component C3 into C3a and C3b. The C3b then binds to the surface of the pathogen and can form additional C3 Convertases, leading to a positive feedback loop that results in the rapid accumulation of complement components on the surface of the pathogen.

This accumulation of complement components helps to mark the pathogen for destruction by other immune cells, such as neutrophils and macrophages. Additionally, the cleavage products C3a and C5a generated during this process can act as anaphylatoxins, inducing inflammation and attracting more immune cells to the site of infection.

Regulation of Complement C3 Convertase is critical to prevent damage to host tissues. Several regulatory proteins, such as factor H and decay-accelerating factor (DAF), help to limit the formation and activity of C3 Convertase on host cells and tissues. Dysregulation of the complement system, including the alternative pathway and Complement C3 Convertase, has been implicated in a variety of diseases, including autoimmune disorders, inflammatory diseases, and infectious diseases.

Complement C3a is a protein fragment that is generated during the activation of the complement system, which is a part of the immune system. The complement system helps to eliminate pathogens and damaged cells from the body by marking them for destruction and attracting immune cells to the site of infection or injury.

C3a is produced when the third component of the complement system (C3) is cleaved into two smaller fragments, C3a and C3b, during the complement activation cascade. C3a is a potent anaphylatoxin, which means it can cause the release of histamine and other mediators from mast cells and basophils, leading to inflammation, increased vascular permeability, and smooth muscle contraction.

C3a also has chemotactic properties, meaning it can attract immune cells such as neutrophils and monocytes to the site of complement activation. Additionally, C3a can modulate the activity of various immune cells, including dendritic cells, T cells, and B cells, and play a role in the regulation of the adaptive immune response.

It's important to note that while C3a has important functions in the immune response, uncontrolled or excessive activation of the complement system can lead to tissue damage and inflammation, contributing to the pathogenesis of various diseases such as autoimmune disorders, inflammatory diseases, and allergies.

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.

Complement C1q is a protein that is part of the complement system, which is a group of proteins in the blood that help to eliminate pathogens and damaged cells from the body. C1q is the first component of the classical complement pathway, which is activated by the binding of C1q to antibodies that are attached to the surface of a pathogen or damaged cell.

C1q is composed of six identical polypeptide chains, each containing a collagen-like region and a globular head region. The globular heads can bind to various structures, including the Fc regions of certain antibodies, immune complexes, and some types of cells. When C1q binds to an activating surface, it triggers a series of proteolytic reactions that lead to the activation of other complement components and the formation of the membrane attack complex (MAC), which can punch holes in the membranes of pathogens or damaged cells, leading to their destruction.

In addition to its role in the immune system, C1q has also been found to have roles in various physiological processes, including tissue remodeling, angiogenesis, and the clearance of apoptotic cells. Dysregulation of the complement system, including abnormalities in C1q function, has been implicated in a variety of diseases, including autoimmune disorders, inflammatory diseases, and neurodegenerative conditions.

The "Classical Complement Pathway" is one of the three pathways that make up the complement system, which is a part of the immune system in humans and other animals. The complement system helps to enhance the ability of antibodies and phagocytic cells to clear pathogens from the body.

The Classical Complement Pathway is initiated by the binding of the first component of the complement system, C1, to an activator surface, such as an antigen-antibody complex. Activation of C1 results in the sequential activation of other components of the complement system, including C4 and C2, which form the C3 convertase (C4b2a). The C3 convertase cleaves the third component of the complement system, C3, into C3a and C3b. C3b then binds to the activator surface and forms a complex with other components of the complement system, leading to the formation of the membrane attack complex (MAC), which creates a pore in the membrane of the target cell, causing its lysis.

The Classical Complement Pathway plays an important role in the immune response to pathogens and can also contribute to inflammation and tissue damage in certain diseases, such as autoimmune disorders and allergies.

Complement C4b is a protein fragment that is formed during the activation of the complement system, which is a part of the immune system. The complement system helps to eliminate pathogens and damaged cells from the body by tagging them for destruction and attracting immune cells to the site of infection or injury.

C4b is generated when the C4 protein is cleaved into two smaller fragments, C4a and C4b, during the activation of the classical or lectin pathways of the complement system. C4b then binds covalently to the surface of the target cell or pathogen, forming a complex with other complement proteins that can create a membrane attack complex (MAC) and cause cell lysis.

C4b can also act as an opsonin, coating the surface of the target cell or pathogen and making it easier for immune cells to recognize and phagocytose them. Additionally, C4b can activate the alternative pathway of the complement system, leading to further amplification of the complement response.

Proprotein convertase 2 (PCSK2) is a type of enzyme known as a proprotein convertase. It plays a role in the activation of other proteins by cleaving off specific peptide sequences and allowing them to become biologically active. PCSK2 is primarily involved in the processing of hormones and neurotransmitters, including insulin, prolactin, and members of the bombesin family.

Defects in the gene that encodes PCSK2 have been associated with certain medical conditions, such as congenital hyperinsulinism, a disorder characterized by low blood sugar levels due to excessive insulin secretion. However, more research is needed to fully understand the relationship between PCSK2 and these conditions.

Complement C5a is a protein fragment that is generated during the activation of the complement system, which is a part of the immune system. The complement system helps to eliminate pathogens and damaged cells from the body by tagging them for destruction and attracting immune cells to the site of infection or injury.

C5a is formed when the fifth component of the complement system (C5) is cleaved into two smaller fragments, C5a and C5b, during the complement activation cascade. C5a is a potent pro-inflammatory mediator that can attract and activate various immune cells, such as neutrophils, monocytes, and eosinophils, to the site of infection or injury. It can also increase vascular permeability, promote the release of histamine, and induce the production of reactive oxygen species, all of which contribute to the inflammatory response.

However, excessive or uncontrolled activation of the complement system and generation of C5a can lead to tissue damage and inflammation, contributing to the pathogenesis of various diseases, such as sepsis, acute respiratory distress syndrome (ARDS), and autoimmune disorders. Therefore, targeting C5a or its receptors has been explored as a potential therapeutic strategy for these conditions.

Complement C2 is a protein that plays a crucial role in the complement system, which is a part of the immune system that helps to eliminate pathogens and damaged cells from the body. Specifically, C2 is a component of the classical complement pathway, which is activated by the binding of antibodies to antigens on the surface of foreign particles or cells.

When the classical pathway is activated, C2 is cleaved into two fragments: C2a and C2b. C2a then binds to C4b to form the C3 convertase (C4b2a), which cleaves C3 into C3a and C3b. C3b can then go on to form the membrane attack complex, which creates a pore in the membrane of the target cell, leading to its lysis.

In summary, Complement C2 is a protein that helps to activate the complement system and destroy foreign particles or cells through the formation of the C3 convertase and the membrane attack complex.

Proprotein convertase 1 (PCSK1) is a protein-coding gene that encodes for the prohormone convertase 1/3 (PC1/3), also known as PCsk1 or PCSK1. This enzyme belongs to the family of subtilisin-like proprotein convertases, which play crucial roles in processing and activating various peptide hormones and neuropeptides by cleaving their precursor proteins.

PC1/3 is primarily expressed in neuroendocrine cells, neurons, and enteroendocrine cells of the gastrointestinal tract. It is involved in the maturation of several bioactive peptides, such as:

1. Proinsulin: PC1/3 processes proinsulin into insulin and C-peptide.
2. Proglucagon: PC1/3 cleaves proglucagon to generate glucagon-like peptide-1 (GLP-1), glucagon-like peptide-2 (GLP-2), glicentin, and oxyntomodulin.
3. Proopiomelanocortin (POMC): PC1/3 processes POMC to generate adrenocorticotropic hormone (ACTH), β-lipotropin, β-endorphin, and melanocyte-stimulating hormones (MSH).
4. Prohormone convertase 2 (PCSK2) precursor: PC1/3 cleaves the PCSK2 precursor into its mature form.
5. Neuropeptide YY (NPY): PC1/3 processes NPY precursors to generate NPY and peptide YY (PYY).
6. Proghrelin: PC1/3 converts proghrelin into acylated ghrelin, which stimulates appetite, and desacyl ghrelin, which has no known function.

Defects in the PCSK1 gene can lead to various endocrine disorders, such as monogenic forms of diabetes (MODY), obesity, and short stature.

Complement activating enzymes are proteins that play a crucial role in the activation of the complement system, which is a part of the immune system. The complement system is a complex series of biochemical reactions that help to eliminate pathogens and damaged cells from the body.

There are several types of complement activating enzymes, including:

1. Classical pathway activators: These include the C1, C4, and C2 components of the complement system. When activated, they trigger a series of reactions that lead to the formation of the membrane attack complex (MAC), which creates a pore in the membrane of the target cell, leading to its lysis.
2. Alternative pathway activators: These include factors B, D, and P. They are constantly active at low levels and can be activated by surfaces that are not normally found in the body, such as bacterial cell walls. Once activated, they also trigger the formation of the MAC.
3. Lectin pathway activators: These include mannose-binding lectin (MBL) and ficolins. They bind to carbohydrates on the surface of microbes and activate the complement system through the MBL-associated serine proteases (MASPs).

Overall, complement activating enzymes play a critical role in the immune response by helping to identify and eliminate pathogens and damaged cells from the body.

Complement inactivator proteins are a group of regulatory proteins that help to control and limit the activation of the complement system, which is a part of the immune system. The complement system is a complex series of biochemical reactions that help to eliminate pathogens and damaged cells from the body. However, if not properly regulated, the complement system can also cause damage to healthy tissues and contribute to the development of various diseases.

Complement inactivator proteins work by inhibiting specific components of the complement system, preventing them from activating and causing an immune response. Some examples of complement inactivator proteins include:

1. C1 inhibitor (C1INH): This protein regulates the activation of the classical pathway of the complement system by inhibiting the C1 complex, which is a group of proteins that initiate this pathway.
2. Decay-accelerating factor (DAF or CD55): This protein regulates the activation of both the classical and alternative pathways of the complement system by accelerating the decay of the C3/C5 convertases, which are enzymes that activate the complement components C3 and C5.
3. Membrane cofactor protein (MCP or CD46): This protein regulates the activation of the alternative pathway of the complement system by serving as a cofactor for the cleavage and inactivation of C3b, a component of the C3 convertase.
4. Factor H: This protein also regulates the activation of the alternative pathway of the complement system by acting as a cofactor for the cleavage and inactivation of C3b, and by preventing the formation of the C3 convertase.

Deficiencies or dysfunction of complement inactivator proteins can lead to various diseases, including hereditary angioedema (C1INH deficiency), atypical hemolytic uremic syndrome (factor H deficiency or dysfunction), and age-related macular degeneration (complement component overactivation).

Properdin is defined as a positive regulatory protein in the complement system, which is a part of the immune system. It plays a crucial role in the alternative pathway of complement activation. Properdin stabilizes the C3 convertase (C3bBb), preventing its decay and increasing the efficiency of the alternative pathway. This results in the production of the membrane attack complex, which leads to the lysis of foreign cells or pathogens. Deficiencies in properdin can lead to an increased susceptibility to bacterial infections.

Complement C6 is a protein that plays a crucial role in the complement system, which is a part of the immune system that helps to eliminate pathogens and damaged cells from the body. Specifically, C6 is a component of the membrane attack complex (MAC), which is a group of proteins that work together to form a pore in the membrane of target cells, leading to their lysis or destruction.

The complement system is activated through several different pathways, including the classical pathway, the lectin pathway, and the alternative pathway. Once activated, these pathways converge at the level of C3, which is cleaved into C3a and C3b fragments. C3b can then bind to the surface of target cells and initiate the formation of the MAC.

C6 is one of several proteins that are required for the formation of the MAC. When C6 binds to C7, it undergoes a conformational change that allows it to interact with C8 and form a stable complex. This complex then recruits additional C9 molecules, which polymerize to form the pore in the target cell membrane.

Deficiencies in complement components, including C6, can lead to increased susceptibility to certain types of infections, as well as autoimmune disorders and other medical conditions.

Complement Factor H is a protein involved in the regulation of the complement system, which is a part of the immune system that helps to clear pathogens and damaged cells from the body. Specifically, Complement Factor H helps to regulate the activation and deactivation of the complement component C3b, preventing excessive or unwanted activation of the complement system and protecting host tissues from damage.

Complement Factor H is a crucial protein in maintaining the balance between the protective effects of the complement system and the potential for harm to the body's own cells and tissues. Deficiencies or mutations in Complement Factor H have been associated with several diseases, including age-related macular degeneration (AMD), atypical hemolytic uremic syndrome (aHUS), and C3 glomerulopathy.

Complement C3d is a protein fragment that is formed during the activation of the complement system, which is a part of the immune system. The complement system helps to eliminate pathogens such as bacteria and viruses from the body by tagging them for destruction and attracting immune cells to the site of infection.

C3d is a cleavage product of complement component C3, which is one of the central proteins in the complement system. When C3 is activated, it is cleaved into two fragments: C3a and C3b. C3b can then be further cleaved into C3d and C3c.

C3d plays a role in the activation of the immune system by helping to link the complement system with the adaptive immune response. It does this by binding to receptors on B cells, which are a type of white blood cell that produces antibodies. This interaction can help to stimulate the production of antibodies and enhance the immune response to pathogens.

C3d has also been implicated in the development of certain autoimmune diseases, as it can contribute to the formation of immune complexes that can cause tissue damage.

Complement Factor D is a protein that plays a crucial role in the complement system, which is a part of the immune system that helps to clear pathogens and damaged cells from the body. Specifically, Factor D is a serine protease that is involved in the alternative pathway of the complement system.

In this pathway, Factor D helps to cleave another protein called Factor B, which then activates a complex called the C3 convertase. The C3 convertase cleaves complement component 3 (C3) into C3a and C3b, leading to the formation of the membrane attack complex (MAC), which creates a pore in the membrane of the target cell, causing its lysis and removal from the body.

Deficiencies or mutations in Complement Factor D can lead to an impaired alternative pathway and increased susceptibility to certain infections, particularly those caused by Neisseria bacteria. Additionally, abnormal regulation of the complement system has been implicated in a variety of diseases, including autoimmune disorders, inflammatory conditions, and neurodegenerative diseases.

Complement C3c is a protein component of the complement system, which is a part of the immune system that helps to clear pathogens and damaged cells from the body. Complement C3c is formed when the third component of the complement system (C3) is cleaved into two smaller proteins, C3a and C3b, during the complement activation process.

C3b can then be further cleaved into C3c and C3dg. C3c is a stable fragment that remains in the circulation and can be measured in blood tests as a marker of complement activation. It plays a role in the opsonization of pathogens, which means it coats them to make them more recognizable to immune cells, and helps to initiate the membrane attack complex (MAC), which forms a pore in the cell membrane of pathogens leading to their lysis or destruction.

Abnormal levels of C3c may indicate an underlying inflammatory or immune-mediated condition, such as infection, autoimmune disease, or cancer.

Complement C9 is a protein that plays a crucial role in the complement system, which is a part of the immune system that helps to eliminate pathogens and damaged cells from the body. Specifically, C9 is one of the components of the membrane attack complex (MAC), which is a protein structure that forms pores in the membranes of target cells, leading to their lysis or destruction.

When activated, C9 polymerizes and inserts itself into the cell membrane, forming a transmembrane pore that disrupts the membrane's integrity and causes the cell to lyse. This process is an essential part of the complement system's ability to destroy pathogens and clear damaged cells from the body.

Defects in the C9 gene can lead to a rare genetic disorder called complement component 9 deficiency, which is characterized by recurrent bacterial infections and immune complex-mediated diseases. Additionally, mutations in the C9 gene have been associated with an increased risk of age-related macular degeneration (AMD), a leading cause of blindness in older adults.

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

There are several types of complement receptors, including:

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

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

Proprotein convertase 5 (PC5, also known as PCSK5 or PACE4) is a serine protease enzyme that belongs to the family of proprotein convertases. These enzymes play crucial roles in processing and activating various protein precursors by cleaving them at specific recognition sites.

PC5 is primarily involved in the activation of other proteins through proteolytic processing, which means it cuts large protein precursors into their smaller, active forms. It has a wide range of substrates, including hormones, growth factors, receptors, and adhesion molecules. PC5 is synthesized as an inactive zymogen and undergoes autocatalytic activation to become fully functional.

PC5 is expressed in various tissues, such as the brain, pancreas, testis, ovary, and placenta. Its dysregulation has been implicated in several diseases, including cancer, neurodegenerative disorders, and viral infections. However, more research is needed to fully understand its functions and therapeutic potential.

Proprotein convertases (PCs) are a group of calcium-dependent serine proteases that play a crucial role in the post-translational modification of proteins. They are responsible for cleaving proproteins into their active forms by removing the propeptide or inhibitory sequences, thereby regulating various biological processes such as protein maturation, activation, and trafficking.

There are nine known human proprotein convertases, including PC1/3, PC2, PC4, PACE4, PC5/6, PC7, Furin, Subtilisin/Kexin type 1 Protease (SKI-1/S1P), and Neuropsin. These enzymes are characterized by their conserved catalytic domain and a distinct prodomain that regulates their activity.

Proprotein convertases have been implicated in several physiological processes, including blood coagulation, neuroendocrine signaling, immune response, and cell differentiation. Dysregulation of these enzymes has been associated with various diseases, such as cancer, cardiovascular disorders, neurological disorders, and infectious diseases. Therefore, understanding the function and regulation of proprotein convertases is essential for developing novel therapeutic strategies to target these diseases.

The Complement Membrane Attack Complex (MAC), also known as the Terminal Complement Complex (TCC), is a protein structure that forms in the final stages of the complement system's immune response. The complement system is a part of the innate immune system that helps to eliminate pathogens and damaged cells from the body.

The MAC is composed of several proteins, including C5b, C6, C7, C8, and multiple subunits of C9, which assemble on the surface of target cells. The formation of the MAC creates a pore-like structure in the cell membrane, leading to disruption of the membrane's integrity and ultimately causing cell lysis or damage.

The MAC plays an important role in the immune response by helping to eliminate pathogens that have evaded other immune defenses. However, uncontrolled activation of the complement system and formation of the MAC can also contribute to tissue damage and inflammation in various diseases, such as autoimmune disorders, age-related macular degeneration, and ischemia-reperfusion injury.

Complement C1s is a protein that plays a crucial role in the complement system, which is a part of the immune system that helps to clear pathogens and damaged cells from the body. Specifically, C1s is a component of the first protein complex in the classical complement pathway, called C1.

C1 is composed of three subunits: C1q, C1r, and C1s. When C1 encounters an activating surface, such as an antibody-antigen complex or certain types of viruses and bacteria, it undergoes a conformational change that allows C1r to cleave and activate C1s. Activated C1s then goes on to cleave and activate other components in the complement pathway, leading to the generation of the membrane attack complex (MAC) and subsequent lysis of the target cell.

Deficiencies or mutations in the genes encoding complement proteins, including C1s, can lead to various immune disorders and increased susceptibility to infections.

Complement C3b inactivator proteins, also known as complement regulators or decay-accelerating factor (DAF), are a group of proteins that play a crucial role in regulating the complement system. The complement system is a part of the immune system that helps to eliminate pathogens and damaged cells from the body.

The complement C3b inactivator proteins include two main types: complement receptor 1 (CR1) and decay-accelerating factor (DAF). These proteins work by regulating the formation of the membrane attack complex (MAC), a protein structure that forms pores in the cell membrane, leading to cell lysis.

Complement C3b inactivator proteins bind to C3b and C4b components of the complement system, preventing them from forming the MAC. By doing so, they help to prevent excessive activation of the complement system, which can damage healthy cells and tissues.

Deficiencies or dysfunction of complement C3b inactivator proteins have been associated with several diseases, including autoimmune disorders, inflammatory diseases, and infectious diseases. Therefore, understanding the role of these proteins in regulating the complement system is essential for developing new therapies to treat these conditions.

Complement C7 is a protein that plays a role in the complement system, which is a part of the immune system that helps to clear pathogens and damaged cells from the body. Specifically, C7 is a component of the membrane attack complex (MAC), which is a group of proteins that forms a pore in the membrane of target cells, leading to their lysis or destruction.

C7 is activated when it binds to the C5b-7 complex, which is formed by the cleavage of C5 and C6 by the C5 convertase. Once bound to the C5b-7 complex, C7 undergoes a conformational change that allows it to insert into the target cell membrane. This forms the basis for the formation of the MAC and subsequent lysis of the target cell.

Deficiencies in complement components, including C7, can lead to increased susceptibility to certain infections and autoimmune disorders. Additionally, abnormal regulation of the complement system has been implicated in a variety of diseases, including inflammatory and degenerative conditions.

Complement C1 is a protein complex that plays a crucial role in the complement system, which is a part of the immune system that helps to eliminate pathogens and damaged cells from the body. The complement system consists of a group of proteins that work together to destroy microbes and remove debris.

Complement C1 is composed of three subunits: C1q, C1r, and C1s. When activated, C1q binds to the surface of a pathogen or damaged cell, leading to the activation of C1r and C1s. Activated C1r then cleaves and activates C1s, which in turn cleaves and activates other complement components, ultimately resulting in the formation of the membrane attack complex (MAC), a protein structure that forms a pore in the membrane of the target cell, leading to its lysis and destruction.

Defects in the complement component C1 can lead to immune disorders, such as hereditary angioedema, which is characterized by recurrent episodes of swelling in various parts of the body.

Complement receptor 3b (CR3b or CD11b/CD18) is not a medical definition itself, but I can provide you with the relevant information regarding this term.

Complement receptor 3 (CR3) is a heterodimeric receptor consisting of two subunits, CD11b (also known as Mac-1 or CR3 alpha) and CD18 (also known as beta2 integrin). There are two forms of the CD11b/CD18 heterodimer: CR3a (CD11b/CD18) and CR3b (CD11b/CD18'). The difference between these two forms lies in the conformation of the CD11b subunit.

Complement receptor 3b (CR3b or CD11b/CD18') is a less common form of the CR3 receptor, which is primarily expressed on myeloid cells such as monocytes, macrophages, and neutrophils. CR3b has a higher affinity for complement component C3b and its fragments iC3b and C3dg compared to CR3a.

CR3b plays a role in various immune functions, including:

1. Phagocytosis: Binding of C3b or its fragments to CR3b facilitates the recognition and uptake of opsonized pathogens by phagocytes.
2. Adhesion: The integrin component of CR3b mediates cell-cell and cell-matrix interactions, contributing to leukocyte migration and recruitment to sites of inflammation or infection.
3. Intracellular signaling: Activation of CR3b can lead to intracellular signaling events that modulate immune responses, such as the release of pro-inflammatory cytokines and reactive oxygen species.

In summary, Complement receptor 3b (CR3b or CD11b/CD18') is a less common form of CR3 primarily expressed on myeloid cells that binds complement component C3b and its fragments with high affinity, mediating phagocytosis, adhesion, and intracellular signaling.

Complement C3 Nephritic Factor (C3NeF) is a type of autoantibody that activates the complement system and plays a significant role in the development of certain types of kidney diseases. The complement system is a part of the immune system that helps to eliminate pathogens and damaged cells from the body.

C3NeF is specifically directed against the C3 convertase enzyme complex, which is a critical component of the complement system's activation pathway. By binding to this enzyme complex, C3NeF stabilizes it and enhances its activity, leading to excessive complement activation and subsequent tissue damage.

In the context of kidney diseases, C3NeF has been associated with several forms of glomerulonephritis, including membranoproliferative glomerulonephritis (MPGN) type II, also known as dense deposit disease (DDD). The persistent activation of the complement system by C3NeF can result in the accumulation of complement components and immune complexes in the glomeruli, causing inflammation, tissue injury, and ultimately leading to kidney function impairment.

It is essential to diagnose and monitor C3NeF levels in patients with kidney diseases, as it may help guide treatment decisions and assess disease prognosis. Therapeutic strategies targeting the complement system, such as eculizumab, have shown promising results in managing C3NeF-associated kidney diseases.

Complement C1r is a protein that plays a crucial role in the complement system, which is a part of the immune system that helps to clear pathogens and damaged cells from the body. Specifically, C1r is one of the three proteins that make up the C1 complex, which is the first component of the classical complement pathway.

The C1 complex is composed of C1q, C1r, and C1s, and it is activated by the binding of C1q to the Fc region of an antibody that is bound to a pathogen or damaged cell. Once activated, C1r undergoes a conformational change that allows it to cleave and activate C1s. Activated C1s then goes on to cleave and activate other components of the complement system, leading to the production of the membrane attack complex (MAC), which forms a pore in the membrane of the target cell and causes lysis.

Deficiencies or mutations in the genes encoding the proteins of the C1 complex can lead to immune disorders, including hereditary angioedema, which is characterized by recurrent episodes of swelling in various parts of the body.

Complement C5b is a protein complex that forms during the activation of the complement system, which is a part of the immune system. The complement system helps to eliminate pathogens and damaged cells from the body by marking them for destruction and attracting immune cells to the site of infection or injury.

The complement component C5 is cleaved into two fragments, C5a and C5b, during the activation of the complement system. C5a is a small peptide that acts as a chemoattractant, drawing immune cells to the site of inflammation. C5b, on the other hand, forms a complex with other complement components (C6, C7, C8, and C9) to create the membrane attack complex (MAC). The MAC inserts itself into the membrane of the target cell, forming a pore that disrupts the cell's integrity and leads to its lysis or destruction.

Therefore, Complement C5b is an important protein involved in the immune response, specifically in the terminal phase of complement activation, which results in the formation of the MAC and subsequent destruction of target cells.

Subtilisins are a group of serine proteases that are produced by certain bacteria, including Bacillus subtilis. They are named after the bacterium and the Latin word "subtilis," which means delicate or finely made. Subtilisins are alkaline proteases, meaning they work best in slightly basic conditions.

Subtilisins have a broad specificity for cleaving peptide bonds and can hydrolyze a wide range of protein substrates. They are widely used in industry for various applications such as detergents, food processing, leather treatment, and biotechnology due to their ability to function at high temperatures and in the presence of denaturing agents.

In medicine, subtilisins have been studied for their potential use in therapeutic applications, including as anti-inflammatory agents and in wound healing. However, more research is needed to fully understand their mechanisms of action and potential benefits.

Furin is not a medical condition or disease, but rather it is a type of enzyme that belongs to the group of proteases. It's also known as paired basic amino acid cleaving enzyme (PACE) or convertase 6.

Furin plays an essential role in processing and activating various proteins in the body, particularly those involved in cell signaling, growth regulation, and viral infectivity. Furin works by cutting or cleaving specific amino acid sequences in proteins, allowing them to become active and perform their functions.

In a medical context, furin is often discussed in relation to its role in activating certain viruses, such as HIV, influenza, and coronaviruses (including SARS-CoV-2). Inhibiting furin activity has been explored as a potential therapeutic strategy for treating these viral infections.

Complement C8 is a protein component of the complement system, which is a part of the immune system that helps to eliminate pathogens and damaged cells from the body. Specifically, C8 is a part of the membrane attack complex (MAC), which forms a pore in the membrane of target cells, leading to their lysis or destruction.

C8 is composed of three subunits: alpha, beta, and gamma. It is activated when it binds to the complement component C5b67 complex on the surface of a target cell. Once activated, C8 undergoes a conformational change that allows it to insert into the target cell membrane and form a pore, which disrupts the cell's membrane integrity and can lead to its death.

Deficiencies in complement components, including C8, can make individuals more susceptible to certain infections and autoimmune diseases. Additionally, mutations in the genes encoding complement proteins have been associated with various inherited disorders, such as atypical hemolytic uremic syndrome (aHUS), which is characterized by thrombotic microangiopathy and kidney failure.

CD55, also known as Decay-accelerating factor (DAF), is a protein that acts as an inhibitor of the complement system, which is a part of the immune system. It prevents the formation of the membrane attack complex (MAC) on host cells and tissues, thereby protecting them from damage caused by the complement activation. CD55 is found on the surface of many types of cells in the body, including red blood cells, white blood cells, and cells lining the blood vessels.

As an antigen, CD55 is a molecule that can be recognized by the immune system and stimulate an immune response. However, unlike some other antigens, CD55 does not typically elicit a strong immune response because it is a self-antigen, meaning it is normally present in the body and should not be targeted by the immune system.

In certain medical conditions, such as autoimmune disorders or transplant rejection, the immune system may mistakenly attack cells expressing CD55. In these cases, measuring the levels of CD55 antigens can provide valuable diagnostic information and help guide treatment decisions.

Complement C2a is a proteolytic fragment generated through the activation of the complement component C2 during the classical complement pathway. It is a serine protease that plays a crucial role in the formation of the C3 convertase (C4b2a) complex, which cleaves and activates the complement component C3 into C3a and C3b. This activation step is essential for the initiation of the immune response and elimination of pathogens through various effector mechanisms such as opsonization, phagocytosis, and formation of the membrane attack complex (MAC). Dysregulation or deficiency in complement components, including C2a, can lead to immunological disorders and increased susceptibility to infections.

Alternative splicing is a process in molecular biology that occurs during the post-transcriptional modification of pre-messenger RNA (pre-mRNA) molecules. It involves the removal of non-coding sequences, known as introns, and the joining together of coding sequences, or exons, to form a mature messenger RNA (mRNA) molecule that can be translated into a protein.

In alternative splicing, different combinations of exons are selected and joined together to create multiple distinct mRNA transcripts from a single pre-mRNA template. This process increases the diversity of proteins that can be produced from a limited number of genes, allowing for greater functional complexity in organisms.

Alternative splicing is regulated by various cis-acting elements and trans-acting factors that bind to specific sequences in the pre-mRNA molecule and influence which exons are included or excluded during splicing. Abnormal alternative splicing has been implicated in several human diseases, including cancer, neurological disorders, and cardiovascular disease.

Complement Factor I is a protein involved in the regulation of the complement system, which is a part of the immune system that helps to clear pathogens and damaged cells from the body. Specifically, Complement Factor I is a serine protease that regulates the complement component C3b by cleaving it into inactive fragments, thereby preventing the excessive activation of the complement system and protecting host tissues from damage.

Complement Factor I functions in conjunction with other regulatory proteins, such as complement receptor 1 (CR1) and membrane cofactor protein (MCP), to control the activity of the complement system at various stages. Deficiencies or mutations in Complement Factor I have been associated with several diseases, including atypical hemolytic uremic syndrome (aHUS), age-related macular degeneration (AMD), and systemic lupus erythematosus (SLE).

The complement system is a part of the immune system that helps to eliminate pathogens and damaged cells from the body. The alternative pathway is one of three pathways that can activate the complement system. Complement C3-C5 convertases are proteins that play a crucial role in the activation of the complement system through the alternative pathway.

Complement C3-C5 convertases are formed by the cleavage of complement component C3 into C3a and C3b by the C3 convertase enzyme. The C3b fragment then binds to the surface of a pathogen or damaged cell, forming a new complex called the C3bBb convertase. This complex can then cleave additional C3 molecules into C3a and C3b fragments, leading to the amplification of the complement response.

The C3bBb convertase can also cleave complement component C5 into C5a and C5b fragments. The C5b fragment then forms a complex with other complement components to form the membrane attack complex (MAC), which creates a pore in the membrane of the pathogen or damaged cell, leading to its lysis and elimination from the body.

Therefore, Complement C3-C5 convertases, Alternative Pathway refer to the enzyme complexes that play a critical role in the activation of the complement system through the alternative pathway, ultimately leading to the destruction of pathogens or damaged cells.

Complement inactivating agents are substances or drugs that inhibit the complement system, which is a part of the immune system responsible for the recognition and elimination of foreign substances and microorganisms. The complement system consists of a group of proteins that work together to help eliminate pathogens from the body.

Complement inactivating agents are used in medical settings to prevent or treat various conditions associated with excessive or unwanted activation of the complement system, such as inflammation, autoimmune diseases, and transplant rejection. These agents can inhibit different components of the complement pathway, including C1 esterase inhibitors, C3 convertase inhibitors, and C5a receptor antagonists.

Examples of complement inactivating agents include eculizumab, ravulizumab, and Alexion's Ultomiris, which are monoclonal antibodies that target C5, a protein involved in the final steps of the complement pathway. These drugs have been approved for the treatment of paroxysmal nocturnal hemoglobinuria (PNH), atypical hemolytic uremic syndrome (aHUS), and other complement-mediated diseases.

Other complement inactivating agents include C1 esterase inhibitors, such as Berinert and Ruconest, which are used to treat hereditary angioedema (HAE). These drugs work by inhibiting the activation of the classical pathway of the complement system, thereby preventing the release of inflammatory mediators that can cause swelling and pain.

Overall, complement inactivating agents play an important role in the treatment of various complement-mediated diseases, helping to reduce inflammation, prevent tissue damage, and improve patient outcomes.

Hemolysis is the destruction or breakdown of red blood cells, resulting in the release of hemoglobin into the surrounding fluid (plasma). This process can occur due to various reasons such as chemical agents, infections, autoimmune disorders, mechanical trauma, or genetic abnormalities. Hemolysis may lead to anemia and jaundice, among other complications. It is essential to monitor hemolysis levels in patients undergoing medical treatments that might cause this condition.

A Complement Hemolytic Activity Assay is a laboratory test used to measure the functionality and activity level of the complement system, which is a part of the immune system. The complement system is a group of proteins that work together to help eliminate pathogens from the body.

The assay measures the ability of the complement system to lyse (break open) red blood cells. This is done by mixing the patient's serum (the liquid portion of the blood) with antibody-coated red blood cells and incubating them together. The complement proteins in the serum will then bind to the antibodies on the red blood cells and cause them to lyse.

The degree of hemolysis (red blood cell lysis) is directly proportional to the activity level of the complement system. By measuring the amount of hemolysis, the assay can determine whether the complement system is functioning properly and at what level of activity.

This test is often used to diagnose or monitor complement-mediated diseases such as autoimmune disorders, infections, and some types of cancer. It may also be used to evaluate the effectiveness of treatments that target the complement system.

Complement C5 Convertase, Alternative Pathway is a protein complex that plays a crucial role in the alternative pathway of the complement system, which is a part of the immune system that helps to eliminate pathogens and damaged cells. The complement system consists of a group of proteins that work together to help recognize and destroy foreign substances, such as bacteria and viruses.

The alternative pathway is one of three pathways that can activate the complement system, and it is constantly active at low levels in the body. The C5 convertase enzyme is formed by the cleavage of a protein called C3 into two fragments, C3a and C3b. The C3b fragment then binds to another protein called factor B, which is activated by a protease called factor D to form the C3bBb complex, known as the C5 convertase of the alternative pathway.

The C5 convertase cleaves the complement protein C5 into two fragments, C5a and C5b. The C5a fragment acts as a chemoattractant, recruiting immune cells to the site of infection or injury. The C5b fragment, along with other complement proteins, forms the membrane attack complex (MAC), which creates a pore in the membrane of the target cell, leading to its lysis and death.

Regulation of the alternative pathway is critical to prevent damage to host cells and tissues. Several regulatory proteins, such as factor H and factor I, help to control the activation of the C5 convertase and limit its activity to prevent excessive complement activation. Dysregulation of the complement system has been implicated in various diseases, including autoimmune disorders, inflammatory conditions, and neurodegenerative diseases.

Complement receptor 3d (CR3d or CD11b/CD18) is not a medical definition in itself, but rather a specific type of integrin receptor that plays a crucial role in the immune system. Here's a breakdown of the components:

1. Complement Receptors: These are proteins found on the surface of various cells, including white blood cells (leukocytes), that recognize and bind to complement components, which are proteins involved in the immune response. The binding of complement components to their receptors helps facilitate communication between cells, enhances phagocytosis (the process by which certain cells engulf and destroy foreign particles or microorganisms), and contributes to the inflammatory response.
2. CR3 (Complement Receptor 3): Complement Receptor 3 is a heterodimeric receptor composed of two subunits, CD11b (also known as integrin alpha M) and CD18 (also known as integrin beta 2). Together, they form the integrin Mac-1 or αMβ2.
3. CR3d (CD11b/CD18): CR3d specifically refers to the CD11b subunit of the Complement Receptor 3 heterodimer. The CD11b subunit is responsible for recognizing and binding to complement component C3b, iC3b, and C4b fragments, as well as other ligands such as fibrinogen, ICAM-1 (Intercellular Adhesion Molecule 1), and factor X.

In summary, Complement Receptor 3d (CR3d or CD11b/CD18) is a type of integrin receptor found on the surface of various immune cells that recognizes and binds to complement components C3b, iC3b, and C4b fragments, as well as other ligands. This binding facilitates communication between cells, enhances phagocytosis, and contributes to the inflammatory response.

Cobra venoms are a type of snake venom that is produced by cobras, which are members of the genus Naja in the family Elapidae. These venoms are complex mixtures of proteins and other molecules that have evolved to help the snake immobilize and digest its prey.

Cobra venoms typically contain a variety of toxic components, including neurotoxins, hemotoxins, and cytotoxins. Neurotoxins target the nervous system and can cause paralysis and respiratory failure. Hemotoxins damage blood vessels and tissues, leading to internal bleeding and organ damage. Cytotoxins destroy cells and can cause tissue necrosis.

The specific composition of cobra venoms can vary widely between different species of cobras, as well as between individual snakes of the same species. Some cobras have venoms that are primarily neurotoxic, while others have venoms that are more hemotoxic or cytotoxic. The potency and effects of cobra venoms can also be influenced by factors such as the age and size of the snake, as well as the temperature and pH of the environment.

Cobra bites can be extremely dangerous and even fatal to humans, depending on the species of cobra, the amount of venom injected, and the location of the bite. Immediate medical attention is required in the event of a cobra bite, including the administration of antivenom therapy to neutralize the effects of the venom.

The term "Receptor, Anaphylatoxin C5a" refers to a specific type of receptor found on the surface of various cells in the human body, including immune cells and endothelial cells. This receptor binds to a molecule called C5a, which is a cleavage product of the complement component C5 and is one of the most potent anaphylatoxins.

Anaphylatoxins are inflammatory mediators that play a crucial role in the immune response, particularly in the activation of the complement system and the recruitment of immune cells to sites of infection or injury. C5a is generated during the activation of the complement system and has a wide range of biological activities, including chemotaxis (attracting immune cells to the site of inflammation), increased vascular permeability, and the activation of immune cells such as neutrophils, monocytes, and mast cells.

The C5a receptor, also known as CD88, is a G protein-coupled receptor that belongs to the superfamily of seven transmembrane domain receptors. When C5a binds to the receptor, it triggers a series of intracellular signaling events that lead to the activation of various cellular responses, such as the release of inflammatory mediators and the recruitment of immune cells to the site of inflammation.

Abnormal activation of the C5a/C5a receptor pathway has been implicated in a variety of inflammatory diseases, including sepsis, acute respiratory distress syndrome (ARDS), and autoimmune disorders. Therefore, targeting this pathway with therapeutic agents has emerged as a promising strategy for the treatment of these conditions.

Complement C4b-binding protein (C4bp) is a regulatory protein in the complement system, which is a part of the immune system that helps to clear pathogens and damaged cells from the body. C4bp regulates the complement system by binding to and inhibiting the activity of C4b, an activated component of the classical and lectin pathways of the complement system. By doing so, C4bp helps to prevent excessive or inappropriate activation of the complement system, which could otherwise lead to tissue damage and inflammation.

C4bp is a complex protein that consists of several subunits, including a central α-chain and multiple β-chains. It is produced by liver cells and can also be found on the surface of some cells in the body. Mutations in the genes encoding C4bp have been associated with certain immune disorders, such as systemic lupus erythematosus (SLE) and atypical hemolytic uremic syndrome (aHUS).

Anaphylatoxins are a group of small protein molecules that are released during an immune response, specifically as a result of the activation of the complement system. The term "anaphylatoxin" comes from their ability to induce anaphylaxis, a severe and rapid allergic reaction. There are three main anaphylatoxins, known as C3a, C4a, and C5a, which are derived from the cleavage of complement components C3, C4, and C5, respectively.

Anaphylatoxins play a crucial role in the immune response by attracting and activating various immune cells, such as neutrophils, eosinophils, and mast cells, to the site of infection or injury. They also increase vascular permeability, causing fluid to leak out of blood vessels and leading to tissue swelling. Additionally, anaphylatoxins can induce smooth muscle contraction, which can result in bronchoconstriction and hypotension.

While anaphylatoxins are important for the immune response, they can also contribute to the pathogenesis of various inflammatory diseases, such as asthma, arthritis, and sepsis. Therefore, therapies that target the complement system and anaphylatoxin production have been developed and are being investigated as potential treatments for these conditions.

Complement C2b is a proteolytic fragment generated through the activation of the complement component C2, which is a part of the complement system. The complement system is a group of plasma proteins that play an important role in the body's immune defense against pathogens and abnormal cells.

When the complement component C2 is activated by the C3 convertase (a complex formed by C3b and either C4b or C4d), it is cleaved into two fragments: C2a and C2b. The C2b fragment is a smaller piece that contains an active protease domain, which can cleave other proteins in the complement pathway.

C2b plays a role in the formation of the membrane attack complex (MAC), a protein structure that forms pores in the membranes of target cells, leading to their lysis and removal by the immune system. Dysregulation or overactivation of the complement system, including C2b, can contribute to various pathological conditions such as autoimmune diseases, inflammatory disorders, and tissue damage.

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.

Complement C1 Inactivator proteins are a part of the complement system, which is a group of proteins in the blood that play a crucial role in the body's immune defense system. Specifically, Complement C1 Inactivator proteins are responsible for regulating the activation of the first component of the complement system, C1.

The complement system is activated in response to the presence of foreign substances such as bacteria or viruses in the body. The activation of C1 leads to a cascade of reactions that result in the destruction of the foreign substance. However, if this process is not properly regulated, it can lead to damage to the body's own cells and tissues.

Complement C1 Inactivator proteins help to prevent this by regulating the activation of C1. They do this by binding to and inhibiting the activity of C1, preventing it from initiating the complement cascade. A deficiency in Complement C1 Inactivator proteins can lead to a condition called hereditary angioedema, which is characterized by recurrent episodes of swelling in various parts of the body.

Complement C3 Convertase, Classical Pathway is a protein complex that plays a crucial role in the activation of the complement system, which is a part of the immune system. The complement system helps to eliminate pathogens and damaged cells from the body.

The classical pathway is one of three pathways that can activate the complement system, with the other two being the lectin pathway and the alternative pathway. The classical pathway is initiated by the binding of antibodies to antigens on the surface of a pathogen or damaged cell. This binding event triggers a series of proteolytic cleavage reactions that result in the formation of the C3 Convertase complex.

The C3 Convertase complex, specifically the C4b2a form, is composed of two proteins: C4b and C2a. These proteins are derived from the cleavage of complement components C4 and C2 by an enzyme called C1 esterase. The C3 Convertase complex then cleaves complement component C3 into C3a and C3b fragments, which initiate further downstream reactions in the complement cascade.

The formation of the C3 Convertase complex is a critical step in the activation of the classical pathway, as it marks the point at which the complement system begins to amplify its response and recruit other immune cells to help eliminate the target pathogen or damaged cell. Dysregulation of the complement system, including abnormalities in the formation or function of the C3 Convertase complex, can contribute to a variety of diseases, including autoimmune disorders, inflammatory conditions, and infections.

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.

Neuroendocrine Secretory Protein 7B2 (NESP7B2) is defined as a protein that is encoded by the 7B2 gene in humans. This protein is primarily produced in neuroendocrine cells, including those found in the brain and the endocrine system. NESP7B2 has a molecular weight of approximately 29 kDa and is composed of 256 amino acids.

One of the primary functions of NESP7B2 is to regulate the activity of another protein called prohormone convertase 2 (PC2). PC2 is involved in the processing and activation of various hormones and neurotransmitters, and NESP7B2 helps to control its activity by binding to it and inhibiting its action.

NESP7B2 has also been found to have a role in the regulation of calcium homeostasis and may be involved in the development and function of the nervous system. Mutations in the 7B2 gene have been associated with certain medical conditions, including some forms of cancer and neurological disorders.

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

There are two main types of opsonins:

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

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

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.

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.

Complement C5 Convertase, Classical Pathway is a protein complex that plays a crucial role in the activation of the complement system, which is a part of the immune system. The complement system helps to eliminate pathogens and damaged cells from the body.

The classical pathway is one of three pathways that can activate the complement system, with the other two being the lectin pathway and the alternative pathway. The activation of the classical pathway begins when an antibody binds to a pathogen or foreign substance. This binding event causes the activation of the C1 complex, which is made up of three proteins: C1q, C1r, and C1s.

Once activated, the C1 complex cleaves C4 and C2 proteins into smaller fragments, C4a, C4b, C2a, and C2b. The C4b and C2a fragments then combine to form the C3 convertase of the classical pathway, also known as C4b2a. This protein complex can cleave C3 protein into C3a and C3b fragments.

The C3b fragment can then bind to the surface of the pathogen or foreign substance, leading to the formation of the membrane attack complex (MAC), which creates a pore in the cell membrane and ultimately leads to the lysis of the target cell. The complement system is tightly regulated to prevent damage to host cells, and deficiencies in any of the components can lead to increased susceptibility to infections.

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

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

Complement fixation tests are a type of laboratory test used in immunology and serology to detect the presence of antibodies in a patient's serum. These tests are based on the principle of complement activation, which is a part of the immune response. The complement system consists of a group of proteins that work together to help eliminate pathogens from the body.

In a complement fixation test, the patient's serum is mixed with a known antigen and complement proteins. If the patient has antibodies against the antigen, they will bind to it and activate the complement system. This results in the consumption or "fixation" of the complement proteins, which are no longer available to participate in a secondary reaction.

A second step involves adding a fresh source of complement proteins and a dye-labeled antibody that recognizes a specific component of the complement system. If complement was fixed during the first step, it will not be available for this secondary reaction, and the dye-labeled antibody will remain unbound. Conversely, if no antibodies were present in the patient's serum, the complement proteins would still be available for the second reaction, leading to the binding of the dye-labeled antibody.

The mixture is then examined under a microscope or using a spectrophotometer to determine whether the dye-labeled antibody has bound. If it has not, this indicates that the patient's serum contains antibodies specific to the antigen used in the test, and a positive result is recorded.

Complement fixation tests have been widely used for the diagnosis of various infectious diseases, such as syphilis, measles, and influenza. However, they have largely been replaced by more modern serological techniques, like enzyme-linked immunosorbent assays (ELISAs) and nucleic acid amplification tests (NAATs), due to their increased sensitivity, specificity, and ease of use.

CD59 is a type of protein found on the surface of many cells in the human body, including red and white blood cells, that functions as an inhibitor of the complement system. The complement system is a part of the immune system that helps to eliminate pathogens such as bacteria and viruses from the body.

CD59 specifically inhibits the formation of the membrane attack complex (MAC), which is a protein structure that forms pores in the cell membrane and can lead to cell lysis or death. By preventing the formation of the MAC, CD59 helps to protect cells from complement-mediated damage.

As an antigen, CD59 is a molecule that can be recognized by the immune system and stimulate an immune response. However, because it is a self-protein found on normal human cells, CD59 is not typically targeted by the immune system unless there is some kind of dysregulation or abnormality.

In certain medical conditions, such as autoimmune disorders or transplant rejection, the immune system may mistakenly target CD59 or other self-proteins, leading to damage to healthy cells and tissues. In these cases, treatments may be necessary to modulate or suppress the immune response and prevent further harm.

The Mannose-Binding Lectin (MBL) pathway is a part of the complement system, which is a group of proteins that play a crucial role in the body's immune defense against infectious agents. The MBL pathway is an alternative activation pathway of the complement system, which can be initiated without the need for antibodies.

MBL is a protein found in blood plasma and other bodily fluids. It recognizes and binds to specific sugars (mannose and fucose) found on the surface of many microorganisms, including bacteria, viruses, fungi, and parasites. When MBL binds to these sugars, it triggers a series of proteolytic cleavage events that activate the complement components C4 and C2, forming the C3 convertase (C4b2a).

The C3 convertase then cleaves the complement component C3 into C3a and C3b. C3b can bind to the surface of microorganisms, leading to their opsonization (coating) and subsequent phagocytosis by immune cells. Additionally, C3b can also trigger the formation of the membrane attack complex (MAC), which creates a pore in the membrane of microorganisms, leading to their lysis and death.

Overall, the MBL pathway plays an essential role in innate immunity, providing a rapid and effective defense against invading microorganisms.

CD46, also known as membrane cofactor protein (MCP), is a regulatory protein that plays a role in the immune system and helps to protect cells from complement activation. It is found on the surface of many different types of cells in the body, including cells of the immune system such as T cells and B cells, as well as cells of various other tissues such as epithelial cells and endothelial cells.

As an antigen, CD46 is a molecule that can be recognized by the immune system and stimulate an immune response. It is a type I transmembrane protein that consists of four distinct domains: two short cytoplasmic domains, a transmembrane domain, and a large extracellular domain. The extracellular domain contains several binding sites for complement proteins, which helps to regulate the activation of the complement system and prevent it from damaging host cells.

CD46 has been shown to play a role in protecting cells from complement-mediated damage, modulating immune responses, and promoting the survival and proliferation of certain types of immune cells. It is also thought to be involved in the development of some autoimmune diseases and may be a target for immunotherapy in the treatment of cancer.

Carboxypeptidase H is also known as carboxypeptidase E or CPE. It is an enzyme that plays a role in the processing and activation of neuropeptides, which are small protein-like molecules that function as chemical messengers within the nervous system. Carboxypeptidase H/E is responsible for removing certain amino acids from the end of newly synthesized neuropeptides, allowing them to become biologically active. It is widely expressed in the brain and other tissues throughout the body.

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.

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.

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.

The Complement C1 Inhibitor protein, also known as C1-INH, is a protein involved in the regulation of the complement system and the contact system, which are parts of the immune system. The complement system helps to eliminate pathogens (e.g., bacteria, viruses) from the body, while the contact system helps to regulate blood coagulation and inflammation.

C1-INH works by inhibiting the activation of C1, an enzyme complex that is the first component of the classical complement pathway. By inhibiting C1, C1-INH prevents the activation of downstream components of the complement system, thereby helping to regulate the immune response and prevent excessive inflammation.

Deficiencies or dysfunction in the C1-INH protein can lead to a group of genetic disorders known as C1 inhibitor deficiency disorders, which include hereditary angioedema (HAE) and acquired angioedema (AAE). These conditions are characterized by recurrent episodes of swelling in various parts of the body, such as the face, hands, feet, and airway, which can be painful and potentially life-threatening if they affect the airway.

Membranoproliferative Glomerulonephritis (MPGN) is a type of glomerulonephritis, which is a group of kidney disorders characterized by inflammation and damage to the glomeruli, the tiny blood vessels in the kidneys responsible for filtering waste and excess fluids from the blood.

MPGN is specifically characterized by thickening of the glomerular basement membrane and proliferation (increased number) of cells in the mesangium, a region within the glomerulus. This condition can be primary or secondary to other diseases such as infections, autoimmune disorders, or monoclonal gammopathies.

MPGN is typically classified into three types based on the pattern of injury seen on electron microscopy: Type I, Type II (Dense Deposit Disease), and Type III. Each type has distinct clinical features, laboratory findings, and treatment approaches. Symptoms of MPGN may include hematuria (blood in urine), proteinuria (protein in urine), hypertension (high blood pressure), edema (swelling), and eventually progress to chronic kidney disease or end-stage renal disease if left untreated.

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.

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

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

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

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.

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.

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.

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.

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.

Complement C3-C5 convertases are protease complexes that play a crucial role in the classical pathway of the complement system, which is a part of the immune system that helps to eliminate pathogens and damaged cells from the body.

The complement system consists of a group of plasma proteins that work together to help eliminate foreign substances and initiate the inflammatory response. The classical pathway is initiated when an antibody binds to a pathogen or antigen, forming an antigen-antibody complex. This complex then activates the first protein in the classical pathway, C1, which subsequently activates the next protein, C4, and then C2.

The resulting complex of C4 and C2 (C4b2a) is called the C3 convertase of the classical pathway. This enzyme cleaves the complement component C3 into C3a and C3b. The C3b fragment then binds to the surface of the pathogen or antigen, forming a new complex with other complement components (C3bBb), which is called the C5 convertase of the classical pathway.

The C5 convertase cleaves the complement component C5 into C5a and C5b. The C5b fragment then initiates the formation of the membrane attack complex (MAC), which creates a pore in the membrane of the pathogen or cell, leading to its lysis and elimination.

Therefore, the Complement C3-C5 convertases, Classical Pathway are essential components of the classical pathway of the complement system that help to eliminate pathogens and damaged cells from the body by activating the complement system and initiating the membrane attack complex.

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.

Enzyme precursors are typically referred to as zymogens or proenzymes. These are inactive forms of enzymes that can be activated under specific conditions. When the need for the enzyme's function arises, the proenzyme is converted into its active form through a process called proteolysis, where it is cleaved by another enzyme. This mechanism helps control and regulate the activation of certain enzymes in the body, preventing unwanted or premature reactions. A well-known example of an enzyme precursor is trypsinogen, which is converted into its active form, trypsin, in the digestive system.

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.

Blood proteins, also known as serum proteins, are a group of complex molecules present in the blood that are essential for various physiological functions. These proteins include albumin, globulins (alpha, beta, and gamma), and fibrinogen. They play crucial roles in maintaining oncotic pressure, transporting hormones, enzymes, vitamins, and minerals, providing immune defense, and contributing to blood clotting.

Albumin is the most abundant protein in the blood, accounting for about 60% of the total protein mass. It functions as a transporter of various substances, such as hormones, fatty acids, and drugs, and helps maintain oncotic pressure, which is essential for fluid balance between the blood vessels and surrounding tissues.

Globulins are divided into three main categories: alpha, beta, and gamma globulins. Alpha and beta globulins consist of transport proteins like lipoproteins, hormone-binding proteins, and enzymes. Gamma globulins, also known as immunoglobulins or antibodies, are essential for the immune system's defense against pathogens.

Fibrinogen is a protein involved in blood clotting. When an injury occurs, fibrinogen is converted into fibrin, which forms a mesh to trap platelets and form a clot, preventing excessive bleeding.

Abnormal levels of these proteins can indicate various medical conditions, such as liver or kidney disease, malnutrition, infections, inflammation, or autoimmune disorders. Blood protein levels are typically measured through laboratory tests like serum protein electrophoresis (SPE) and immunoelectrophoresis (IEP).

Blood bactericidal activity refers to the ability of an individual's blood to kill or inhibit the growth of bacteria. This is an important aspect of the body's immune system, as it helps to prevent infection and maintain overall health. The bactericidal activity of blood can be influenced by various factors, including the presence of antibodies, white blood cells (such as neutrophils), and complement proteins.

In medical terms, the term "bactericidal" specifically refers to an agent or substance that is capable of killing bacteria. Therefore, when we talk about blood bactericidal activity, we are referring to the collective ability of various components in the blood to kill or inhibit the growth of bacteria. This is often measured in laboratory tests as a way to assess a person's immune function and their susceptibility to infection.

It's worth noting that not all substances in the blood are bactericidal; some may simply inhibit the growth of bacteria without killing them. These substances are referred to as bacteriostatic. Both bactericidal and bacteriostatic agents play important roles in maintaining the body's defense against infection.

"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.

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.

Protein precursors, also known as proproteins or prohormones, are inactive forms of proteins that undergo post-translational modification to become active. These modifications typically include cleavage of the precursor protein by specific enzymes, resulting in the release of the active protein. This process allows for the regulation and control of protein activity within the body. Protein precursors can be found in various biological processes, including the endocrine system where they serve as inactive hormones that can be converted into their active forms when needed.

Tertiary protein structure refers to the three-dimensional arrangement of all the elements (polypeptide chains) of a single protein molecule. It is the highest level of structural organization and results from interactions between various side chains (R groups) of the amino acids that make up the protein. These interactions, which include hydrogen bonds, ionic bonds, van der Waals forces, and disulfide bridges, give the protein its unique shape and stability, which in turn determines its function. The tertiary structure of a protein can be stabilized by various factors such as temperature, pH, and the presence of certain ions. Any changes in these factors can lead to denaturation, where the protein loses its tertiary structure and thus its function.

Glomerulonephritis is a medical condition that involves inflammation of the glomeruli, which are the tiny blood vessel clusters in the kidneys that filter waste and excess fluids from the blood. This inflammation can impair the kidney's ability to filter blood properly, leading to symptoms such as proteinuria (protein in the urine), hematuria (blood in the urine), edema (swelling), hypertension (high blood pressure), and eventually kidney failure.

Glomerulonephritis can be acute or chronic, and it may occur as a primary kidney disease or secondary to other medical conditions such as infections, autoimmune disorders, or vasculitis. The diagnosis of glomerulonephritis typically involves a combination of medical history, physical examination, urinalysis, blood tests, and imaging studies, with confirmation often requiring a kidney biopsy. Treatment depends on the underlying cause and severity of the disease but may include medications to suppress inflammation, control blood pressure, and manage symptoms.

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.

Post-translational protein processing refers to the modifications and changes that proteins undergo after their synthesis on ribosomes, which are complex molecular machines responsible for protein synthesis. These modifications occur through various biochemical processes and play a crucial role in determining the final structure, function, and stability of the protein.

The process begins with the translation of messenger RNA (mRNA) into a linear polypeptide chain, which is then subjected to several post-translational modifications. These modifications can include:

1. Proteolytic cleavage: The removal of specific segments or domains from the polypeptide chain by proteases, resulting in the formation of mature, functional protein subunits.
2. Chemical modifications: Addition or modification of chemical groups to the side chains of amino acids, such as phosphorylation (addition of a phosphate group), glycosylation (addition of sugar moieties), methylation (addition of a methyl group), acetylation (addition of an acetyl group), and ubiquitination (addition of a ubiquitin protein).
3. Disulfide bond formation: The oxidation of specific cysteine residues within the polypeptide chain, leading to the formation of disulfide bonds between them. This process helps stabilize the three-dimensional structure of proteins, particularly in extracellular environments.
4. Folding and assembly: The acquisition of a specific three-dimensional conformation by the polypeptide chain, which is essential for its function. Chaperone proteins assist in this process to ensure proper folding and prevent aggregation.
5. Protein targeting: The directed transport of proteins to their appropriate cellular locations, such as the nucleus, mitochondria, endoplasmic reticulum, or plasma membrane. This is often facilitated by specific signal sequences within the protein that are recognized and bound by transport machinery.

Collectively, these post-translational modifications contribute to the functional diversity of proteins in living organisms, allowing them to perform a wide range of cellular processes, including signaling, catalysis, regulation, and structural support.

Mannose-binding protein-associated serine proteases (MASPs) are a group of enzymes that are associated with mannose-binding lectin (MBL), a protein involved in the innate immune system's response to pathogens. MASPs are responsible for activating the complement system, which is a part of the immune system that helps to eliminate pathogens and damaged cells from the body.

MASPs are proteases, meaning they cleave other proteins at specific sites. There are two main types of MASPs, MASP-1 and MASP-2, which are activated by the binding of MBL to carbohydrate structures on the surface of pathogens. Once activated, MASP-1 and MASP-2 cleave complement components C4 and C2, leading to the formation of the C3 convertase enzyme complex, which ultimately results in the activation of the complement system.

MASPs have also been shown to play a role in other physiological processes, such as tissue remodeling and inflammation. Mutations in MASP genes have been associated with various immune disorders, including recurrent infections, autoimmune diseases, and inflammatory conditions.

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.

Steroid 21-hydroxylase, also known as CYP21A2, is a crucial enzyme involved in the synthesis of steroid hormones in the adrenal gland. Specifically, it catalyzes the conversion of 17-hydroxyprogesterone to 11-deoxycortisol and progesterone to deoxycorticosterone in the glucocorticoid and mineralocorticoid pathways, respectively.

Deficiency or mutations in this enzyme can lead to a group of genetic disorders called congenital adrenal hyperplasia (CAH), which is characterized by impaired cortisol production and disrupted hormonal balance. Depending on the severity of the deficiency, CAH can result in various symptoms such as ambiguous genitalia, precocious puberty, sexual infantilism, infertility, and increased risk of adrenal crisis.

Mannose-Binding Lectin (MBL) is a protein that belongs to the collectin family and plays a crucial role in the innate immune system. It's primarily produced by the liver and secreted into the bloodstream. MBL binds to carbohydrate structures, such as mannose, found on the surface of various microorganisms, including bacteria, viruses, fungi, and parasites.

Once MBL binds to these microorganisms, it activates the complement system through the lectin pathway, which leads to the destruction of the pathogens by opsonization (marking for phagocytosis) or direct lysis. Additionally, MBL can also initiate other immune responses, such as inflammation and immune cell activation, helping to protect the host from infections.

Deficiencies in MBL have been associated with increased susceptibility to certain infectious diseases, autoimmune disorders, and allergies. However, more research is needed to fully understand the complex role of MBL in human health and disease.

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.

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.

Paroxysmal nocturnal hemoglobinuria (PNH) is a rare, acquired disorder of the blood characterized by the destruction of red blood cells (hemolysis), which can cause symptoms such as fatigue, dark colored urine (especially in the morning), chest pain, shortness of breath, and an increased risk of blood clots. The hemoglobin from the lysed red blood cells appears in the urine, hence the term "hemoglobinuria."

The paroxysmal nature of the disorder refers to the sudden and recurring episodes of hemolysis that can occur at any time, although they may be more frequent at night. The condition is caused by mutations in a gene called PIG-A, which leads to the production of defective red blood cell membranes that are sensitive to destruction by complement, a component of the immune system.

PNH is a serious and potentially life-threatening condition that can lead to complications such as kidney damage, pulmonary hypertension, and thrombosis. Treatment typically involves supportive care, such as blood transfusions, and medications to manage symptoms and prevent complications. In some cases, stem cell transplantation may be considered as a curative treatment option.

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.

Esterases are a group of enzymes that catalyze the hydrolysis of ester bonds in esters, producing alcohols and carboxylic acids. They are widely distributed in plants, animals, and microorganisms and play important roles in various biological processes, such as metabolism, digestion, and detoxification.

Esterases can be classified into several types based on their substrate specificity, including carboxylesterases, cholinesterases, lipases, and phosphatases. These enzymes have different structures and mechanisms of action but all share the ability to hydrolyze esters.

Carboxylesterases are the most abundant and diverse group of esterases, with a wide range of substrate specificity. They play important roles in the metabolism of drugs, xenobiotics, and lipids. Cholinesterases, on the other hand, specifically hydrolyze choline esters, such as acetylcholine, which is an important neurotransmitter in the nervous system. Lipases are a type of esterase that preferentially hydrolyzes triglycerides and plays a crucial role in fat digestion and metabolism. Phosphatases are enzymes that remove phosphate groups from various molecules, including esters, and have important functions in signal transduction and other cellular processes.

Esterases can also be used in industrial applications, such as in the production of biodiesel, detergents, and food additives. They are often produced by microbial fermentation or extracted from plants and animals. The use of esterases in biotechnology is an active area of research, with potential applications in biofuel production, bioremediation, and medical diagnostics.

Sequence homology, amino acid, refers to the similarity in the order of amino acids in a protein or a portion of a protein between two or more species. This similarity can be used to infer evolutionary relationships and functional similarities between proteins. The higher the degree of sequence homology, the more likely it is that the proteins are related and have similar functions. Sequence homology can be determined through various methods such as pairwise alignment or multiple sequence alignment, which compare the sequences and calculate a score based on the number and type of matching amino acids.

Electrophoresis, polyacrylamide gel (EPG) is a laboratory technique used to separate and analyze complex mixtures of proteins or nucleic acids (DNA or RNA) based on their size and electrical charge. This technique utilizes a matrix made of cross-linked polyacrylamide, a type of gel, which provides a stable and uniform environment for the separation of molecules.

In this process:

1. The polyacrylamide gel is prepared by mixing acrylamide monomers with a cross-linking agent (bis-acrylamide) and a catalyst (ammonium persulfate) in the presence of a buffer solution.
2. The gel is then poured into a mold and allowed to polymerize, forming a solid matrix with uniform pore sizes that depend on the concentration of acrylamide used. Higher concentrations result in smaller pores, providing better resolution for separating smaller molecules.
3. Once the gel has set, it is placed in an electrophoresis apparatus containing a buffer solution. Samples containing the mixture of proteins or nucleic acids are loaded into wells on the top of the gel.
4. An electric field is applied across the gel, causing the negatively charged molecules to migrate towards the positive electrode (anode) while positively charged molecules move toward the negative electrode (cathode). The rate of migration depends on the size, charge, and shape of the molecules.
5. Smaller molecules move faster through the gel matrix and will migrate farther from the origin compared to larger molecules, resulting in separation based on size. Proteins and nucleic acids can be selectively stained after electrophoresis to visualize the separated bands.

EPG is widely used in various research fields, including molecular biology, genetics, proteomics, and forensic science, for applications such as protein characterization, DNA fragment analysis, cloning, mutation detection, and quality control of nucleic acid or protein samples.

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.

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.

Serum, in the context of clinical and medical laboratory science, refers to the fluid that is obtained after blood coagulation. It is the yellowish, straw-colored liquid fraction of whole blood that remains after the clotting factors have been removed. Serum contains various proteins, electrolytes, hormones, antibodies, antigens, and other substances, which can be analyzed to help diagnose and monitor a wide range of medical conditions. It is commonly used for various clinical tests such as chemistry panels, immunological assays, drug screening, and infectious disease testing.

Hemolytic-Uremic Syndrome (HUS) is a serious condition that affects the blood and kidneys. It is characterized by three major features: the breakdown of red blood cells (hemolysis), the abnormal clotting of small blood vessels (microthrombosis), and acute kidney failure.

The breakdown of red blood cells leads to the release of hemoglobin into the bloodstream, which can cause anemia. The microthrombi can obstruct the flow of blood in the kidneys' filtering system (glomeruli), leading to damaged kidney function and potentially acute kidney failure.

HUS is often caused by a bacterial infection, most commonly Escherichia coli (E. coli) that produces Shiga toxins. This form of HUS is known as STEC-HUS or Stx-HUS. Other causes include infections with other bacteria, viruses, medications, pregnancy complications, and certain medical conditions such as autoimmune diseases.

Symptoms of HUS may include fever, fatigue, decreased urine output, blood in the stool, swelling in the face, hands, or feet, and irritability or confusion. Treatment typically involves supportive care, including dialysis for kidney failure, transfusions to replace lost red blood cells, and managing high blood pressure. In severe cases, a kidney transplant may be necessary.

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.

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.

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

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

Immunoelectrophoresis (IEP) is a laboratory technique used in the field of clinical pathology and immunology. It is a method for separating and identifying proteins, particularly immunoglobulins or antibodies, in a sample. This technique combines the principles of electrophoresis, which separates proteins based on their electric charge and size, with immunological reactions, which detect specific proteins using antigen-antibody interactions.

In IEP, a protein sample is first separated by electrophoresis in an agarose or agar gel matrix on a glass slide or in a test tube. After separation, an antibody specific to the protein of interest is layered on top of the gel and allowed to diffuse towards the separated proteins. This creates a reaction between the antigen (protein) and the antibody, forming a visible precipitate at the point where they meet. The precipitate line's position and intensity can then be analyzed to identify and quantify the protein of interest.

Immunoelectrophoresis is particularly useful in diagnosing various medical conditions, such as immunodeficiency disorders, monoclonal gammopathies (like multiple myeloma), and other plasma cell dyscrasias. It can help detect abnormal protein patterns, quantify specific immunoglobulins, and identify the presence of M-proteins or Bence Jones proteins, which are indicative of monoclonal gammopathies.

Edetic acid, also known as ethylenediaminetetraacetic acid (EDTA), is not a medical term per se, but a chemical compound with various applications in medicine. EDTA is a synthetic amino acid that acts as a chelating agent, which means it can bind to metallic ions and form stable complexes.

In medicine, EDTA is primarily used in the treatment of heavy metal poisoning, such as lead or mercury toxicity. It works by binding to the toxic metal ions in the body, forming a stable compound that can be excreted through urine. This helps reduce the levels of harmful metals in the body and alleviate their toxic effects.

EDTA is also used in some diagnostic tests, such as the determination of calcium levels in blood. Additionally, it has been explored as a potential therapy for conditions like atherosclerosis and Alzheimer's disease, although its efficacy in these areas remains controversial and unproven.

It is important to note that EDTA should only be administered under medical supervision due to its potential side effects and the need for careful monitoring of its use.

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.

A kidney glomerulus is a functional unit in the nephron of the kidney. It is a tuft of capillaries enclosed within a structure called Bowman's capsule, which filters waste and excess fluids from the blood. The glomerulus receives blood from an afferent arteriole and drains into an efferent arteriole.

The process of filtration in the glomerulus is called ultrafiltration, where the pressure within the glomerular capillaries drives plasma fluid and small molecules (such as ions, glucose, amino acids, and waste products) through the filtration membrane into the Bowman's space. Larger molecules, like proteins and blood cells, are retained in the blood due to their larger size. The filtrate then continues down the nephron for further processing, eventually forming urine.

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.

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.

Aspartic acid endopeptidases are a type of enzyme that cleave peptide bonds within proteins. They are also known as aspartyl proteases or aspartic proteinases. These enzymes contain two catalytic aspartic acid residues in their active site, which work together to hydrolyze the peptide bond.

Aspartic acid endopeptidases play important roles in various biological processes, including protein degradation, processing, and activation. They are found in many organisms, including viruses, bacteria, fungi, plants, and animals. Some well-known examples of aspartic acid endopeptidases include pepsin, cathepsin D, and HIV protease.

Pepsin is a digestive enzyme found in the stomach that helps break down proteins in food. Cathepsin D is a lysosomal enzyme that plays a role in protein turnover and degradation within cells. HIV protease is an essential enzyme for the replication of the human immunodeficiency virus (HIV), which causes AIDS. Inhibitors of HIV protease are used as antiretroviral drugs to treat HIV infection.

Pituitary hormones are chemical messengers produced and released by the pituitary gland, a small endocrine gland located at the base of the brain. The pituitary gland is often referred to as the "master gland" because it controls several other endocrine glands and regulates various bodily functions.

There are two main types of pituitary hormones: anterior pituitary hormones and posterior pituitary hormones, which are produced in different parts of the pituitary gland and have distinct functions.

Anterior pituitary hormones include:

1. Growth hormone (GH): regulates growth and metabolism.
2. Thyroid-stimulating hormone (TSH): stimulates the thyroid gland to produce thyroid hormones.
3. Adrenocorticotropic hormone (ACTH): stimulates the adrenal glands to produce cortisol and other steroid hormones.
4. Follicle-stimulating hormone (FSH) and luteinizing hormone (LH): regulate reproductive function in both males and females.
5. Prolactin: stimulates milk production in lactating women.
6. Melanocyte-stimulating hormone (MSH): regulates skin pigmentation and appetite.

Posterior pituitary hormones include:

1. Oxytocin: stimulates uterine contractions during childbirth and milk ejection during lactation.
2. Vasopressin (antidiuretic hormone, ADH): regulates water balance in the body by controlling urine production in the kidneys.

Overall, pituitary hormones play crucial roles in regulating growth, development, metabolism, reproductive function, and various other bodily functions. Abnormalities in pituitary hormone levels can lead to a range of medical conditions, such as dwarfism, acromegaly, Cushing's disease, infertility, and diabetes insipidus.

Collectins are a group of proteins that belong to the collectin family, which are involved in the innate immune system. They are composed of a collagen-like region and a carbohydrate recognition domain (CRD), which allows them to bind to specific sugars on the surface of microorganisms, cells, and particles. Collectins play a crucial role in the defense against pathogens by promoting the clearance of microbes, modulating inflammation, and regulating immune responses.

Some examples of collectins include:

* Surfactant protein A (SP-A) and surfactant protein D (SP-D), which are found in the lungs and help to maintain the stability of the lung lining and protect against respiratory infections.
* Mannose-binding lectin (MBL), which is a serum protein that binds to mannose sugars on the surface of microorganisms, activating the complement system and promoting phagocytosis.
* Collectin liver 1 (CL-L1) and collectin kidney 1 (CL-K1), which are found in the liver and kidneys, respectively, and play a role in the clearance of apoptotic cells and immune complexes.

Deficiencies or mutations in collectins can lead to increased susceptibility to infections, autoimmune diseases, and other disorders.

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.

Proglucagon is a precursor protein that gets cleaved into several hormones, including glucagon, GLP-1 (Glucagon-like peptide-1), and GLP-2 (Glucagon-like peptide-2). These hormones play crucial roles in regulating blood sugar levels, energy balance, and gut function. Proglucagon is primarily produced by the alpha cells of the pancreas and L cells in the intestine. Glucagon helps to raise blood sugar levels during fasting or hypoglycemia, while GLP-1 and GLP-2 contribute to glucose regulation, satiety, and gut motility, among other functions.

Surface Plasmon Resonance (SPR) is a physical phenomenon that occurs at the interface between a metal and a dielectric material, when electromagnetic radiation (usually light) is shone on it. It involves the collective oscillation of free electrons in the metal, known as surface plasmons, which are excited by the incident light. The resonance condition is met when the momentum and energy of the photons match those of the surface plasmons, leading to a strong absorption of light and an evanescent wave that extends into the dielectric material.

In the context of medical diagnostics and research, SPR is often used as a sensitive and label-free detection technique for biomolecular interactions. By immobilizing one binding partner (e.g., a receptor or antibody) onto the metal surface and flowing the other partner (e.g., a ligand or antigen) over it, changes in the refractive index at the interface can be measured in real-time as the plasmons are disturbed by the presence of bound molecules. This allows for the quantification of binding affinities, kinetics, and specificity with high sensitivity and selectivity.

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.

Exons are the coding regions of DNA that remain in the mature, processed mRNA after the removal of non-coding intronic sequences during RNA splicing. These exons contain the information necessary to encode proteins, as they specify the sequence of amino acids within a polypeptide chain. The arrangement and order of exons can vary between different genes and even between different versions of the same gene (alternative splicing), allowing for the generation of multiple protein isoforms from a single gene. This complexity in exon structure and usage significantly contributes to the diversity and functionality of the proteome.

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).

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.

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.

'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.

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.

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.

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

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

Cricetinae is a subfamily of rodents that includes hamsters, gerbils, and relatives. These small mammals are characterized by having short limbs, compact bodies, and cheek pouches for storing food. They are native to various parts of the world, particularly in Europe, Asia, and Africa. Some species are popular pets due to their small size, easy care, and friendly nature. In a medical context, understanding the biology and behavior of Cricetinae species can be important for individuals who keep them as pets or for researchers studying their physiology.

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.

Pro-opiomelanocortin (POMC) is a precursor protein that gets cleaved into several biologically active peptides in the body. These peptides include adrenocorticotropic hormone (ACTH), beta-lipotropin, and multiple opioid peptides such as beta-endorphin, met-enkephalin, and leu-enkephalin.

ACTH stimulates the release of cortisol from the adrenal gland, while beta-lipotropin has various metabolic functions. The opioid peptides derived from POMC have pain-relieving (analgesic) and rewarding effects in the brain. Dysregulation of the POMC system has been implicated in several medical conditions, including obesity, addiction, and certain types of hormone deficiencies.

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.

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.

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.

CHO cells, or Chinese Hamster Ovary cells, are a type of immortalized cell line that are commonly used in scientific research and biotechnology. They were originally derived from the ovaries of a female Chinese hamster (Cricetulus griseus) in the 1950s.

CHO cells have several characteristics that make them useful for laboratory experiments. They can grow and divide indefinitely under appropriate conditions, which allows researchers to culture large quantities of them for study. Additionally, CHO cells are capable of expressing high levels of recombinant proteins, making them a popular choice for the production of therapeutic drugs, vaccines, and other biologics.

In particular, CHO cells have become a workhorse in the field of biotherapeutics, with many approved monoclonal antibody-based therapies being produced using these cells. The ability to genetically modify CHO cells through various methods has further expanded their utility in research and industrial applications.

It is important to note that while CHO cells are widely used in scientific research, they may not always accurately represent human cell behavior or respond to drugs and other compounds in the same way as human cells do. Therefore, results obtained using CHO cells should be validated in more relevant systems when possible.

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.

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.

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 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.

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

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

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

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

A genetic complementation test is a laboratory procedure used in molecular genetics to determine whether two mutated genes can complement each other's function, indicating that they are located at different loci and represent separate alleles. This test involves introducing a normal or wild-type copy of one gene into a cell containing a mutant version of the same gene, and then observing whether the presence of the normal gene restores the normal function of the mutated gene. If the introduction of the normal gene results in the restoration of the normal phenotype, it suggests that the two genes are located at different loci and can complement each other's function. However, if the introduction of the normal gene does not restore the normal phenotype, it suggests that the two genes are located at the same locus and represent different alleles of the same gene. This test is commonly used to map genes and identify genetic interactions in a variety of organisms, including bacteria, yeast, and animals.

Complementary DNA (cDNA) is a type of DNA that is synthesized from a single-stranded RNA molecule through the process of reverse transcription. In this process, the enzyme reverse transcriptase uses an RNA molecule as a template to synthesize a complementary DNA strand. The resulting cDNA is therefore complementary to the original RNA molecule and is a copy of its coding sequence, but it does not contain non-coding regions such as introns that are present in genomic DNA.

Complementary DNA is often used in molecular biology research to study gene expression, protein function, and other genetic phenomena. For example, cDNA can be used to create cDNA libraries, which are collections of cloned cDNA fragments that represent the expressed genes in a particular cell type or tissue. These libraries can then be screened for specific genes or gene products of interest. Additionally, cDNA can be used to produce recombinant proteins in heterologous expression systems, allowing researchers to study the structure and function of proteins that may be difficult to express or purify from their native sources.

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).

Mannans are a type of complex carbohydrate, specifically a heteropolysaccharide, that are found in the cell walls of certain plants, algae, and fungi. They consist of chains of mannose sugars linked together, often with other sugar molecules such as glucose or galactose.

Mannans have various biological functions, including serving as a source of energy for microorganisms that can break them down. In some cases, mannans can also play a role in the immune response and are used as a component of vaccines to stimulate an immune response.

In the context of medicine, mannans may be relevant in certain conditions such as gut dysbiosis or allergic reactions to foods containing mannans. Additionally, some research has explored the potential use of mannans as a delivery vehicle for drugs or other therapeutic agents.

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.

Biological models, also known as physiological models or organismal models, are simplified representations of biological systems, processes, or mechanisms that are used to understand and explain the underlying principles and relationships. These models can be theoretical (conceptual or mathematical) or physical (such as anatomical models, cell cultures, or animal models). They are widely used in biomedical research to study various phenomena, including disease pathophysiology, drug action, and therapeutic interventions.

Examples of biological models include:

1. Mathematical models: These use mathematical equations and formulas to describe complex biological systems or processes, such as population dynamics, metabolic pathways, or gene regulation networks. They can help predict the behavior of these systems under different conditions and test hypotheses about their underlying mechanisms.
2. Cell cultures: These are collections of cells grown in a controlled environment, typically in a laboratory dish or flask. They can be used to study cellular processes, such as signal transduction, gene expression, or metabolism, and to test the effects of drugs or other treatments on these processes.
3. Animal models: These are living organisms, usually vertebrates like mice, rats, or non-human primates, that are used to study various aspects of human biology and disease. They can provide valuable insights into the pathophysiology of diseases, the mechanisms of drug action, and the safety and efficacy of new therapies.
4. Anatomical models: These are physical representations of biological structures or systems, such as plastic models of organs or tissues, that can be used for educational purposes or to plan surgical procedures. They can also serve as a basis for developing more sophisticated models, such as computer simulations or 3D-printed replicas.

Overall, biological models play a crucial role in advancing our understanding of biology and medicine, helping to identify new targets for therapeutic intervention, develop novel drugs and treatments, and improve human health.

Substrate specificity in the context of medical biochemistry and enzymology refers to the ability of an enzyme to selectively bind and catalyze a chemical reaction with a particular substrate (or a group of similar substrates) while discriminating against other molecules that are not substrates. This specificity arises from the three-dimensional structure of the enzyme, which has evolved to match the shape, charge distribution, and functional groups of its physiological substrate(s).

Substrate specificity is a fundamental property of enzymes that enables them to carry out highly selective chemical transformations in the complex cellular environment. The active site of an enzyme, where the catalysis takes place, has a unique conformation that complements the shape and charge distribution of its substrate(s). This ensures efficient recognition, binding, and conversion of the substrate into the desired product while minimizing unwanted side reactions with other molecules.

Substrate specificity can be categorized as:

1. Absolute specificity: An enzyme that can only act on a single substrate or a very narrow group of structurally related substrates, showing no activity towards any other molecule.
2. Group specificity: An enzyme that prefers to act on a particular functional group or class of compounds but can still accommodate minor structural variations within the substrate.
3. Broad or promiscuous specificity: An enzyme that can act on a wide range of structurally diverse substrates, albeit with varying catalytic efficiencies.

Understanding substrate specificity is crucial for elucidating enzymatic mechanisms, designing drugs that target specific enzymes or pathways, and developing biotechnological applications that rely on the controlled manipulation of enzyme activities.

Two-dimensional immunoelectrophoresis (2DE) is a specialized laboratory technique used in the field of clinical pathology and immunology. This technique is a refined version of traditional immunoelectrophoresis that adds an additional electrophoretic separation step, enhancing its resolution and allowing for more detailed analysis of complex protein mixtures.

In two-dimensional immunoelectrophoresis, proteins are first separated based on their isoelectric points (pI) in the initial dimension using isoelectric focusing (IEF). This process involves applying an electric field to a protein mixture contained within a gel matrix, where proteins will migrate and stop migrating once they reach the pH that matches their own isoelectric point.

Following IEF, the separated proteins are then subjected to a second electrophoretic separation in the perpendicular direction (second dimension) based on their molecular weights using sodium dodecyl sulfate-polyacrylamide gel electrophoresis (SDS-PAGE). SDS is a negatively charged molecule that binds to proteins, giving them a uniform negative charge and allowing for separation based solely on size.

Once the two-dimensional separation is complete, the gel is then overlaid with specific antisera to detect and identify proteins of interest. The resulting precipitin arcs formed at the intersection of the antibody and antigen are compared to known standards or patterns to determine the identity and quantity of the separated proteins.

Two-dimensional immunoelectrophoresis is particularly useful in identifying and quantifying proteins in complex mixtures, such as those found in body fluids like serum, urine, or cerebrospinal fluid (CSF). It can be applied to various clinical scenarios, including diagnosis and monitoring of monoclonal gammopathies, autoimmune disorders, and certain infectious diseases.

Complement C5a, des-Arginine is a derivative of the complement component C5a. The complement system is a group of proteins that are part of the body's immune defense against foreign invaders such as bacteria and viruses. When activated, the complement system can help to eliminate pathogens by attracting immune cells to the site of infection, promoting inflammation, and directly killing the pathogen.

C5a is a small protein that is generated when the complement component C5 is cleaved during the activation of the complement system. C5a is a potent anaphylatoxin, which means it can cause the release of histamine from mast cells and basophils, leading to increased vascular permeability, smooth muscle contraction, and recruitment of immune cells to the site of infection.

Des-Arginine refers to the removal of an arginine residue from the C-terminus of C5a. This modified form of C5a is known as C5a-desArg and has reduced pro-inflammatory activity compared to intact C5a. However, it can still contribute to the regulation of the immune response by interacting with specific receptors on immune cells.

In summary, Complement C5a, des-Arginine is a derivative of the complement component C5a that has reduced pro-inflammatory activity due to the removal of an arginine residue from its C-terminus.

I apologize, but I am not able to provide a medical definition for "Egtazic Acid" because it is not a term that is recognized in the field of medicine or pharmacology. It is possible that you may have meant "Egтарic Acid," which also does not have a specific medical meaning, or "Ethylene Glycol Tetraacetic Acid (EGTA)," which is a chemical compound used in research and medicine for its ability to bind calcium ions. If you have any other questions, I would be happy to try to help answer them.

Nephritis is a medical term that refers to inflammation of the kidneys, specifically affecting the glomeruli - the tiny filtering units inside the kidneys. The condition can cause damage to the glomeruli, leading to impaired kidney function and the leakage of protein and blood into the urine.

Nephritis can result from a variety of causes, including infections, autoimmune disorders, and exposure to certain medications or toxins. Depending on the severity and underlying cause, nephritis may be treated with medications, dietary modifications, or other therapies aimed at reducing inflammation and preserving kidney function. In severe cases, hospitalization and more intensive treatments may be necessary.

Molecular models are three-dimensional representations of molecular structures that are used in the field of molecular biology and chemistry to visualize and understand the spatial arrangement of atoms and bonds within a molecule. These models can be physical or computer-generated and allow researchers to study the shape, size, and behavior of molecules, which is crucial for understanding their function and interactions with other molecules.

Physical molecular models are often made up of balls (representing atoms) connected by rods or sticks (representing bonds). These models can be constructed manually using materials such as plastic or wooden balls and rods, or they can be created using 3D printing technology.

Computer-generated molecular models, on the other hand, are created using specialized software that allows researchers to visualize and manipulate molecular structures in three dimensions. These models can be used to simulate molecular interactions, predict molecular behavior, and design new drugs or chemicals with specific properties. Overall, molecular models play a critical role in advancing our understanding of molecular structures and their functions.

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.

Fibrinogen is a soluble protein present in plasma, synthesized by the liver. It plays an essential role in blood coagulation. When an injury occurs, fibrinogen gets converted into insoluble fibrin by the action of thrombin, forming a fibrin clot that helps to stop bleeding from the injured site. Therefore, fibrinogen is crucial for hemostasis, which is the process of stopping bleeding and starting the healing process after an injury.

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.

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.

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.

Beta-globulins are a group of proteins found in the beta region of a serum protein electrophoresis, which is a laboratory test used to separate and identify different types of proteins in the blood. This group includes several important proteins such as:

1. Beta-lipoproteins: These are responsible for transporting fat molecules, including cholesterol, throughout the body.
2. Transferrin: A protein that binds and transports iron in the blood.
3. Complement components: These proteins play a crucial role in the immune system's response to infection and inflammation.
4. Beta-2 microglobulin: A protein involved in the functioning of the immune system, elevated levels of which can be found in various conditions such as kidney disease and autoimmune disorders.
5. Hemopexin: A protein that binds and transports heme (a component of hemoglobin) in the blood.

It is important to note that any significant increase or decrease in beta-globulins can indicate an underlying medical condition, such as liver disease, kidney disease, or an autoimmune disorder. Therefore, abnormal results should be further evaluated by a healthcare professional for proper diagnosis and treatment.

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.

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.

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.

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.

A kidney, in medical terms, is one of two bean-shaped organs located in the lower back region of the body. They are essential for maintaining homeostasis within the body by performing several crucial functions such as:

1. Regulation of water and electrolyte balance: Kidneys help regulate the amount of water and various electrolytes like sodium, potassium, and calcium in the bloodstream to maintain a stable internal environment.

2. Excretion of waste products: They filter waste products from the blood, including urea (a byproduct of protein metabolism), creatinine (a breakdown product of muscle tissue), and other harmful substances that result from normal cellular functions or external sources like medications and toxins.

3. Endocrine function: Kidneys produce several hormones with important roles in the body, such as erythropoietin (stimulates red blood cell production), renin (regulates blood pressure), and calcitriol (activated form of vitamin D that helps regulate calcium homeostasis).

4. pH balance regulation: Kidneys maintain the proper acid-base balance in the body by excreting either hydrogen ions or bicarbonate ions, depending on whether the blood is too acidic or too alkaline.

5. Blood pressure control: The kidneys play a significant role in regulating blood pressure through the renin-angiotensin-aldosterone system (RAAS), which constricts blood vessels and promotes sodium and water retention to increase blood volume and, consequently, blood pressure.

Anatomically, each kidney is approximately 10-12 cm long, 5-7 cm wide, and 3 cm thick, with a weight of about 120-170 grams. They are surrounded by a protective layer of fat and connected to the urinary system through the renal pelvis, ureters, bladder, and urethra.

Metalloendopeptidases are a type of enzymes that cleave peptide bonds in proteins, specifically at interior positions within the polypeptide chain. They require metal ions as cofactors for their catalytic activity, typically zinc (Zn2+) or cobalt (Co2+). These enzymes play important roles in various biological processes such as protein degradation, processing, and signaling. Examples of metalloendopeptidases include thermolysin, matrix metalloproteinases (MMPs), and neutrophil elastase.

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.

In genetics, sequence alignment is the process of arranging two or more DNA, RNA, or protein sequences to identify regions of similarity or homology between them. This is often done using computational methods to compare the nucleotide or amino acid sequences and identify matching patterns, which can provide insight into evolutionary relationships, functional domains, or potential genetic disorders. The alignment process typically involves adjusting gaps and mismatches in the sequences to maximize the similarity between them, resulting in an aligned sequence that can be visually represented and analyzed.

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.

Oxidoreductases are a class of enzymes that catalyze oxidation-reduction reactions, which involve the transfer of electrons from one molecule (the reductant) to another (the oxidant). These enzymes play a crucial role in various biological processes, including energy production, metabolism, and detoxification.

The oxidoreductase-catalyzed reaction typically involves the donation of electrons from a reducing agent (donor) to an oxidizing agent (acceptor), often through the transfer of hydrogen atoms or hydride ions. The enzyme itself does not undergo any permanent chemical change during this process, but rather acts as a catalyst to lower the activation energy required for the reaction to occur.

Oxidoreductases are classified and named based on the type of electron donor or acceptor involved in the reaction. For example, oxidoreductases that act on the CH-OH group of donors are called dehydrogenases, while those that act on the aldehyde or ketone groups are called oxidases. Other examples include reductases, peroxidases, and catalases.

Understanding the function and regulation of oxidoreductases is important for understanding various physiological processes and developing therapeutic strategies for diseases associated with impaired redox homeostasis, such as cancer, neurodegenerative disorders, and cardiovascular disease.

'Escherichia coli' (E. coli) is a type of gram-negative, facultatively anaerobic, rod-shaped bacterium that commonly inhabits the intestinal tract of humans and warm-blooded animals. It is a member of the family Enterobacteriaceae and one of the most well-studied prokaryotic model organisms in molecular biology.

While most E. coli strains are harmless and even beneficial to their hosts, some serotypes can cause various forms of gastrointestinal and extraintestinal illnesses in humans and animals. These pathogenic strains possess virulence factors that enable them to colonize and damage host tissues, leading to diseases such as diarrhea, urinary tract infections, pneumonia, and sepsis.

E. coli is a versatile organism with remarkable genetic diversity, which allows it to adapt to various environmental niches. It can be found in water, soil, food, and various man-made environments, making it an essential indicator of fecal contamination and a common cause of foodborne illnesses. The study of E. coli has contributed significantly to our understanding of fundamental biological processes, including DNA replication, gene regulation, and protein synthesis.

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

The Northern blotting procedure involves several steps:

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

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

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

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

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

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

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

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

NF-κB (Nuclear Factor kappa-light-chain-enhancer of activated B cells) is a protein complex that regulates many normal cellular and inflammatory responses, including cell survival, differentiation, and apoptosis. NF-κB p52 subunit is one of the several subunits that make up this protein complex.

The p52 subunit is derived from the proteolytic processing of its precursor protein, p100. This process occurs in response to certain stimuli and results in the formation of a mature p52 subunit, which then combines with other NF-κB family members (such as RelB) to form a functional NF-κB heterodimer.

The activated NF-κB complex then translocates to the nucleus, where it binds to specific DNA sequences called κB sites and regulates the expression of target genes involved in various cellular processes, such as immune response, inflammation, differentiation, and stress responses. Dysregulation of NF-κB signaling has been implicated in several diseases, including cancer, autoimmune disorders, and chronic inflammatory conditions.

A Structure-Activity Relationship (SAR) in the context of medicinal chemistry and pharmacology refers to the relationship between the chemical structure of a drug or molecule and its biological activity or effect on a target protein, cell, or organism. SAR studies aim to identify patterns and correlations between structural features of a compound and its ability to interact with a specific biological target, leading to a desired therapeutic response or undesired side effects.

By analyzing the SAR, researchers can optimize the chemical structure of lead compounds to enhance their potency, selectivity, safety, and pharmacokinetic properties, ultimately guiding the design and development of novel drugs with improved efficacy and reduced toxicity.

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.

Endopeptidases are a type of enzyme that breaks down proteins by cleaving peptide bonds inside the polypeptide chain. They are also known as proteinases or endoproteinases. These enzymes work within the interior of the protein molecule, cutting it at specific points along its length, as opposed to exopeptidases, which remove individual amino acids from the ends of the protein chain.

Endopeptidases play a crucial role in various biological processes, such as digestion, blood coagulation, and programmed cell death (apoptosis). They are classified based on their catalytic mechanism and the structure of their active site. Some examples of endopeptidase families include serine proteases, cysteine proteases, aspartic proteases, and metalloproteases.

It is important to note that while endopeptidases are essential for normal physiological functions, they can also contribute to disease processes when their activity is unregulated or misdirected. For instance, excessive endopeptidase activity has been implicated in the pathogenesis of neurodegenerative disorders, cancer, and inflammatory conditions.

Hydrolysis is a chemical process, not a medical one. However, it is relevant to medicine and biology.

Hydrolysis is the breakdown of a chemical compound due to its reaction with water, often resulting in the formation of two or more simpler compounds. In the context of physiology and medicine, hydrolysis is a crucial process in various biological reactions, such as the digestion of food molecules like proteins, carbohydrates, and fats. Enzymes called hydrolases catalyze these hydrolysis reactions to speed up the breakdown process in the body.

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.

Carboxypeptidases are a group of enzymes that catalyze the cleavage of peptide bonds at the carboxyl-terminal end of polypeptides or proteins. They specifically remove the last amino acid residue from the protein chain, provided that it has a free carboxyl group and is not blocked by another chemical group. Carboxypeptidases are classified into two main types based on their catalytic mechanism: serine carboxypeptidases and metallo-carboxypeptidases.

Serine carboxypeptidases, also known as chymotrypsin C or carboxypeptidase C, use a serine residue in their active site to catalyze the hydrolysis of peptide bonds. They are found in various organisms, including animals and bacteria.

Metallo-carboxypeptidases, on the other hand, require a metal ion (usually zinc) for their catalytic activity. They can be further divided into several subtypes based on their structure and substrate specificity. For example, carboxypeptidase A prefers to cleave hydrophobic amino acids from the carboxyl-terminal end of proteins, while carboxypeptidase B specifically removes basic residues (lysine or arginine).

Carboxypeptidases have important roles in various biological processes, such as protein maturation, digestion, and regulation of blood pressure. Dysregulation of these enzymes has been implicated in several diseases, including cancer, neurodegenerative disorders, and cardiovascular disease.

Lentinan is a polysaccharide derived from the shiitake mushroom (Lentinula edodes) that has been studied for its potential immune-enhancing and anti-cancer effects. It is a beta-glucan with a complex structure, including both β-(1,3)-D-glucan and β-(1,6)-D-glucan branches.

In the medical context, lentinan is considered an immunomodulator, as it can stimulate various immune cells like macrophages, neutrophils, and natural killer (NK) cells. These immune cells play crucial roles in recognizing and eliminating foreign substances, including cancer cells and pathogens.

Lentinan has been investigated for its potential therapeutic benefits in treating several types of cancer, such as gastric, colorectal, and breast cancers. However, most of the research is still preclinical or based on small-scale clinical trials, so more extensive studies are needed to confirm its efficacy and safety before it can be widely adopted in medical practice.

It's important to note that lentinan supplements should not replace conventional cancer treatments but may potentially be used as an adjunct therapy under the guidance of a healthcare professional.

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

Glicentin is a polypeptide hormone that is derived from the preproglucagon gene and is secreted by intestinal L cells. It consists of 69 amino acids, including the first 30 amino acids of glucagon and the following 37 amino acids that are unique to glicentin.

Glicentin has several biological activities, including stimulating insulin secretion from pancreatic beta cells, inhibiting gastric acid secretion, and regulating intestinal motility. It also acts as a growth factor for the intestinal mucosa and may have a role in modulating inflammation and immune responses.

Glicentin is often co-secreted with other hormones such as glucagon-like peptide-1 (GLP-1) and GLP-2, which also play important roles in regulating glucose metabolism and intestinal function.

Schistosoma is a genus of flatworms that cause the disease schistosomiasis, also known as snail fever. These parasitic worms infect freshwater snails and then release a form of the parasite that can penetrate the skin of humans when they come into contact with contaminated water. The larvae mature into adult worms in the human body, living in the blood vessels of the bladder, intestines or other organs, where they lay eggs. These eggs can cause serious damage to internal organs and lead to a range of symptoms including fever, chills, diarrhea, and anemia. Schistosomiasis is a significant public health problem in many tropical and subtropical regions around the world.

Animal testing alternatives, also known as alternative methods or replacement methods, refer to scientific techniques that can be used to replace the use of animals in research and testing. These methods aim to achieve the same scientific objectives while avoiding harm to animals. There are several categories of animal testing alternatives:

1. In vitro (test tube or cell culture) methods: These methods involve growing cells or tissues in a laboratory setting, outside of a living organism. They can be used to study the effects of chemicals, drugs, and other substances on specific cell types or tissues.
2. Computer modeling and simulation: Advanced computer programs and algorithms can be used to model biological systems and predict how they will respond to various stimuli. These methods can help researchers understand complex biological processes without using animals.
3. In silico (using computer models) methods: These methods involve the use of computational tools and databases to predict the potential toxicity or other biological effects of chemicals, drugs, and other substances. They can be used to identify potential hazards and prioritize further testing.
4. Microdosing: This method involves giving human volunteers very small doses of a drug or chemical, followed by careful monitoring to assess its safety and pharmacological properties. This approach can provide valuable information while minimizing the use of animals.
5. Tissue engineering: Scientists can create functional tissue constructs using cells, scaffolds, and bioreactors. These engineered tissues can be used to study the effects of drugs, chemicals, and other substances on human tissues without using animals.
6. Human-based approaches: These methods involve the use of human volunteers, donated tissues, or cells obtained from consenting adults. Examples include microdosing, organ-on-a-chip technology, and the use of human cell lines in laboratory experiments.

These animal testing alternatives can help reduce the number of animals used in research and testing, refine experimental procedures to minimize suffering, and replace the use of animals with non-animal methods whenever possible.

Subtilisin is not strictly a medical term, but rather a term used in biochemistry and microbiology. It refers to a group of proteolytic enzymes (proteases) that are produced by certain bacteria, particularly Bacillus subtilis. These enzymes have the ability to break down other proteins into smaller peptides or individual amino acids by cleaving specific peptide bonds.

In a medical context, subtilisin might be mentioned in relation to its use in various commercial products such as detergents and contact lens cleaning solutions, where it helps to break down protein-based stains or deposits. Additionally, subtilisins have been explored for their potential applications in therapeutics, including the treatment of certain diseases caused by protein misfolding or aggregation, like cystic fibrosis and Alzheimer's disease.

However, it is important to note that direct medical definitions of 'subtilisin' are limited, as it primarily functions within the realms of biochemistry and microbiology.

Protein conformation refers to the specific three-dimensional shape that a protein molecule assumes due to the spatial arrangement of its constituent amino acid residues and their associated chemical groups. This complex structure is determined by several factors, including covalent bonds (disulfide bridges), hydrogen bonds, van der Waals forces, and ionic bonds, which help stabilize the protein's unique conformation.

Protein conformations can be broadly classified into two categories: primary, secondary, tertiary, and quaternary structures. The primary structure represents the linear sequence of amino acids in a polypeptide chain. The secondary structure arises from local interactions between adjacent amino acid residues, leading to the formation of recurring motifs such as α-helices and β-sheets. Tertiary structure refers to the overall three-dimensional folding pattern of a single polypeptide chain, while quaternary structure describes the spatial arrangement of multiple folded polypeptide chains (subunits) that interact to form a functional protein complex.

Understanding protein conformation is crucial for elucidating protein function, as the specific three-dimensional shape of a protein directly influences its ability to interact with other molecules, such as ligands, nucleic acids, or other proteins. Any alterations in protein conformation due to genetic mutations, environmental factors, or chemical modifications can lead to loss of function, misfolding, aggregation, and disease states like neurodegenerative disorders and cancer.

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.

Sialic acids are a family of nine-carbon sugars that are commonly found on the outermost surface of many cell types, particularly on the glycoconjugates of mucins in various secretions and on the glycoproteins and glycolipids of cell membranes. They play important roles in a variety of biological processes, including cell recognition, immune response, and viral and bacterial infectivity. Sialic acids can exist in different forms, with N-acetylneuraminic acid being the most common one in humans.

Macular degeneration, also known as age-related macular degeneration (AMD), is a medical condition that affects the central part of the retina, called the macula. The macula is responsible for sharp, detailed vision, which is necessary for activities such as reading, driving, and recognizing faces.

In AMD, there is a breakdown or deterioration of the macula, leading to gradual loss of central vision. There are two main types of AMD: dry (atrophic) and wet (exudative). Dry AMD is more common and progresses more slowly, while wet AMD is less common but can cause rapid and severe vision loss if left untreated.

The exact causes of AMD are not fully understood, but risk factors include age, smoking, family history, high blood pressure, obesity, and exposure to sunlight. While there is no cure for AMD, treatments such as vitamin supplements, laser therapy, and medication injections can help slow its progression and reduce the risk of vision loss.

Magnesium is an essential mineral that plays a crucial role in various biological processes in the human body. It is the fourth most abundant cation in the body and is involved in over 300 enzymatic reactions, including protein synthesis, muscle and nerve function, blood glucose control, and blood pressure regulation. Magnesium also contributes to the structural development of bones and teeth.

In medical terms, magnesium deficiency can lead to several health issues, such as muscle cramps, weakness, heart arrhythmias, and seizures. On the other hand, excessive magnesium levels can cause symptoms like diarrhea, nausea, and muscle weakness. Magnesium supplements or magnesium-rich foods are often recommended to maintain optimal magnesium levels in the body.

Some common dietary sources of magnesium include leafy green vegetables, nuts, seeds, legumes, whole grains, and dairy products. Magnesium is also available in various forms as a dietary supplement, including magnesium oxide, magnesium citrate, magnesium chloride, and magnesium glycinate.

'Azospirillum brasilense' is a species of free-living, nitrogen-fixing bacteria that is commonly found in the soil and in the roots of various plants. It belongs to the genus Azospirillum and is known for its ability to promote plant growth through a process called bacterial colonization. The bacteria colonize the root system of the plant and enhance nutrient uptake, leading to improved growth and yield. Additionally, 'Azospirillum brasilense' can convert atmospheric nitrogen into ammonia, making it available to the plants as a natural fertilizer. It is widely used in agricultural practices as a bioinoculant to improve crop productivity and sustainability.

Arteriolosclerosis is a medical term that refers to the thickening and hardening of the walls of small arteries or arterioles, usually due to the buildup of calcium, fatty deposits, or excessive collagen. This process can lead to decreased blood flow and increased resistance in the affected vessels, potentially causing damage to various organs and contributing to the development of hypertension, kidney disease, and other conditions.

There are two main types of arteriolosclerosis: hyaline arteriolosclerosis and hyperplastic arteriolosclerosis. Hyaline arteriolosclerosis is characterized by the accumulation of a homogeneous, eosinophilic (pink) material called hyaline within the walls of the arterioles. This type of arteriolosclerosis is often associated with aging, diabetes mellitus, and hypertension.

Hyperplastic arteriolosclerosis, on the other hand, involves the proliferation of smooth muscle cells and excessive collagen deposition in the walls of the arterioles. This type of arteriolosclerosis is commonly seen in malignant hypertension and can lead to fibrinoid necrosis, a condition where the vessel wall undergoes degeneration and becomes replaced by fibrin, a protein involved in blood clotting.

In summary, arteriolosclerosis refers to the thickening and hardening of the walls of small arteries or arterioles due to various causes, which can negatively impact organ function and contribute to the development of several medical conditions.

Heparin is defined as a highly sulfated glycosaminoglycan (a type of polysaccharide) that is widely present in many tissues, but is most commonly derived from the mucosal tissues of mammalian lungs or intestinal mucosa. It is an anticoagulant that acts as an inhibitor of several enzymes involved in the blood coagulation cascade, primarily by activating antithrombin III which then neutralizes thrombin and other clotting factors.

Heparin is used medically to prevent and treat thromboembolic disorders such as deep vein thrombosis, pulmonary embolism, and certain types of heart attacks. It can also be used during hemodialysis, cardiac bypass surgery, and other medical procedures to prevent the formation of blood clots.

It's important to note that while heparin is a powerful anticoagulant, it does not have any fibrinolytic activity, meaning it cannot dissolve existing blood clots. Instead, it prevents new clots from forming and stops existing clots from growing larger.

Streptococcus pneumoniae, also known as the pneumococcus, is a gram-positive, alpha-hemolytic bacterium frequently found in the upper respiratory tract of healthy individuals. It is a leading cause of community-acquired pneumonia and can also cause other infectious diseases such as otitis media (ear infection), sinusitis, meningitis, and bacteremia (bloodstream infection). The bacteria are encapsulated, and there are over 90 serotypes based on variations in the capsular polysaccharide. Some serotypes are more virulent or invasive than others, and the polysaccharide composition is crucial for vaccine development. S. pneumoniae infection can be treated with antibiotics, but the emergence of drug-resistant strains has become a significant global health concern.

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.

Immunoglobulin (Ig) Fab fragments are the antigen-binding portions of an antibody that result from the digestion of the whole antibody molecule by enzymes such as papain. An antibody, also known as an immunoglobulin, is a Y-shaped protein produced by the immune system to identify and neutralize foreign substances like bacteria, viruses, or toxins. The antibody has two identical antigen-binding sites, located at the tips of the two shorter arms, which can bind specifically to a target antigen.

Fab fragments are formed when an antibody is cleaved by papain, resulting in two Fab fragments and one Fc fragment. Each Fab fragment contains one antigen-binding site, composed of a variable region (Fv) and a constant region (C). The Fv region is responsible for the specificity and affinity of the antigen binding, while the C region contributes to the effector functions of the antibody.

Fab fragments are often used in various medical applications, such as immunodiagnostics and targeted therapies, due to their ability to bind specifically to target antigens without triggering an immune response or other effector functions associated with the Fc region.

"Nelumbo" is the scientific genus name for the lotus flower, which includes two species: Nelumbo nucifera (also known as Sacred Lotus) and Nelumbo lutea (American Lotus). These aquatic plants are known for their large, beautiful flowers that bloom on the surface of the water. While "Nelumbo" is a term from plant taxonomy and botany, it does not have a specific medical definition as such. However, various parts of Nelumbo plants have been used in traditional medicine across different cultures for treating various health conditions. For instance, the seeds, leaves, and roots of Nelumbo nucifera are used in Traditional Chinese Medicine to treat several ailments like diarrhea, fever, and skin diseases. Nonetheless, it is essential to consult healthcare professionals before using any plant or herbal remedy for medicinal purposes.

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.

Staphylococcus aureus is a type of gram-positive, round (coccal) bacterium that is commonly found on the skin and mucous membranes of warm-blooded animals and humans. It is a facultative anaerobe, which means it can grow in the presence or absence of oxygen.

Staphylococcus aureus is known to cause a wide range of infections, from mild skin infections such as pimples, impetigo, and furuncles (boils) to more severe and potentially life-threatening infections such as pneumonia, endocarditis, osteomyelitis, and sepsis. It can also cause food poisoning and toxic shock syndrome.

The bacterium is often resistant to multiple antibiotics, including methicillin, which has led to the emergence of methicillin-resistant Staphylococcus aureus (MRSA) strains that are difficult to treat. Proper hand hygiene and infection control practices are critical in preventing the spread of Staphylococcus aureus and MRSA.

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.

Gel chromatography is a type of liquid chromatography that separates molecules based on their size or molecular weight. It uses a stationary phase that consists of a gel matrix made up of cross-linked polymers, such as dextran, agarose, or polyacrylamide. The gel matrix contains pores of various sizes, which allow smaller molecules to penetrate deeper into the matrix while larger molecules are excluded.

In gel chromatography, a mixture of molecules is loaded onto the top of the gel column and eluted with a solvent that moves down the column by gravity or pressure. As the sample components move down the column, they interact with the gel matrix and get separated based on their size. Smaller molecules can enter the pores of the gel and take longer to elute, while larger molecules are excluded from the pores and elute more quickly.

Gel chromatography is commonly used to separate and purify proteins, nucleic acids, and other biomolecules based on their size and molecular weight. It is also used in the analysis of polymers, colloids, and other materials with a wide range of applications in chemistry, biology, and medicine.

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

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

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

Solubility is a fundamental concept in pharmaceutical sciences and medicine, which refers to the maximum amount of a substance (solute) that can be dissolved in a given quantity of solvent (usually water) at a specific temperature and pressure. Solubility is typically expressed as mass of solute per volume or mass of solvent (e.g., grams per liter, milligrams per milliliter). The process of dissolving a solute in a solvent results in a homogeneous solution where the solute particles are dispersed uniformly throughout the solvent.

Understanding the solubility of drugs is crucial for their formulation, administration, and therapeutic effectiveness. Drugs with low solubility may not dissolve sufficiently to produce the desired pharmacological effect, while those with high solubility might lead to rapid absorption and short duration of action. Therefore, optimizing drug solubility through various techniques like particle size reduction, salt formation, or solubilization is an essential aspect of drug development and delivery.

"Cricetulus" is a genus of rodents that includes several species of hamsters. These small, burrowing animals are native to Asia and have a body length of about 8-15 centimeters, with a tail that is usually shorter than the body. They are characterized by their large cheek pouches, which they use to store food. Some common species in this genus include the Chinese hamster (Cricetulus griseus) and the Daurian hamster (Cricetulus dauuricus). These animals are often kept as pets or used in laboratory research.

Site-directed mutagenesis is a molecular biology technique used to introduce specific and targeted changes to a specific DNA sequence. This process involves creating a new variant of a gene or a specific region of interest within a DNA molecule by introducing a planned, deliberate change, or mutation, at a predetermined site within the DNA sequence.

The methodology typically involves the use of molecular tools such as PCR (polymerase chain reaction), restriction enzymes, and/or ligases to introduce the desired mutation(s) into a plasmid or other vector containing the target DNA sequence. The resulting modified DNA molecule can then be used to transform host cells, allowing for the production of large quantities of the mutated gene or protein for further study.

Site-directed mutagenesis is a valuable tool in basic research, drug discovery, and biotechnology applications where specific changes to a DNA sequence are required to understand gene function, investigate protein structure/function relationships, or engineer novel biological properties into existing genes or proteins.

Sequence homology in nucleic acids refers to the similarity or identity between the nucleotide sequences of two or more DNA or RNA molecules. It is often used as a measure of biological relationship between genes, organisms, or populations. High sequence homology suggests a recent common ancestry or functional constraint, while low sequence homology may indicate a more distant relationship or different functions.

Nucleic acid sequence homology can be determined by various methods such as pairwise alignment, multiple sequence alignment, and statistical analysis. The degree of homology is typically expressed as a percentage of identical or similar nucleotides in a given window of comparison.

It's important to note that the interpretation of sequence homology depends on the biological context and the evolutionary distance between the sequences compared. Therefore, functional and experimental validation is often necessary to confirm the significance of sequence homology.

Bacterial antibodies are a type of antibodies produced by the immune system in response to an infection caused by bacteria. These antibodies are proteins that recognize and bind to specific antigens on the surface of the bacterial cells, marking them for destruction by other immune cells. Bacterial antibodies can be classified into several types based on their structure and function, including IgG, IgM, IgA, and IgE. They play a crucial role in the body's defense against bacterial infections and provide immunity to future infections with the same bacteria.

COS cells are a type of cell line that are commonly used in molecular biology and genetic research. The name "COS" is an acronym for "CV-1 in Origin," as these cells were originally derived from the African green monkey kidney cell line CV-1. COS cells have been modified through genetic engineering to express high levels of a protein called SV40 large T antigen, which allows them to efficiently take up and replicate exogenous DNA.

There are several different types of COS cells that are commonly used in research, including COS-1, COS-3, and COS-7 cells. These cells are widely used for the production of recombinant proteins, as well as for studies of gene expression, protein localization, and signal transduction.

It is important to note that while COS cells have been a valuable tool in scientific research, they are not without their limitations. For example, because they are derived from monkey kidney cells, there may be differences in the way that human genes are expressed or regulated in these cells compared to human cells. Additionally, because COS cells express SV40 large T antigen, they may have altered cell cycle regulation and other phenotypic changes that could affect experimental results. Therefore, it is important to carefully consider the choice of cell line when designing experiments and interpreting results.

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.

Genotype, in genetics, refers to the complete heritable genetic makeup of an individual organism, including all of its genes. It is the set of instructions contained in an organism's DNA for the development and function of that organism. The genotype is the basis for an individual's inherited traits, and it can be contrasted with an individual's phenotype, which refers to the observable physical or biochemical characteristics of an organism that result from the expression of its genes in combination with environmental influences.

It is important to note that an individual's genotype is not necessarily identical to their genetic sequence. Some genes have multiple forms called alleles, and an individual may inherit different alleles for a given gene from each parent. The combination of alleles that an individual inherits for a particular gene is known as their genotype for that gene.

Understanding an individual's genotype can provide important information about their susceptibility to certain diseases, their response to drugs and other treatments, and their risk of passing on inherited genetic disorders to their offspring.

"Saccharomyces cerevisiae" is not typically considered a medical term, but it is a scientific name used in the field of microbiology. It refers to a species of yeast that is commonly used in various industrial processes, such as baking and brewing. It's also widely used in scientific research due to its genetic tractability and eukaryotic cellular organization.

However, it does have some relevance to medical fields like medicine and nutrition. For example, certain strains of S. cerevisiae are used as probiotics, which can provide health benefits when consumed. They may help support gut health, enhance the immune system, and even assist in the digestion of certain nutrients.

In summary, "Saccharomyces cerevisiae" is a species of yeast with various industrial and potential medical applications.

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.

The brain is the central organ of the nervous system, responsible for receiving and processing sensory information, regulating vital functions, and controlling behavior, movement, and cognition. It is divided into several distinct regions, each with specific functions:

1. Cerebrum: The largest part of the brain, responsible for higher cognitive functions such as thinking, learning, memory, language, and perception. It is divided into two hemispheres, each controlling the opposite side of the body.
2. Cerebellum: Located at the back of the brain, it is responsible for coordinating muscle movements, maintaining balance, and fine-tuning motor skills.
3. Brainstem: Connects the cerebrum and cerebellum to the spinal cord, controlling vital functions such as breathing, heart rate, and blood pressure. It also serves as a relay center for sensory information and motor commands between the brain and the rest of the body.
4. Diencephalon: A region that includes the thalamus (a major sensory relay station) and hypothalamus (regulates hormones, temperature, hunger, thirst, and sleep).
5. Limbic system: A group of structures involved in emotional processing, memory formation, and motivation, including the hippocampus, amygdala, and cingulate gyrus.

The brain is composed of billions of interconnected neurons that communicate through electrical and chemical signals. It is protected by the skull and surrounded by three layers of membranes called meninges, as well as cerebrospinal fluid that provides cushioning and nutrients.

Hydrogen-ion concentration, also known as pH, is a measure of the acidity or basicity of a solution. It is defined as the negative logarithm (to the base 10) of the hydrogen ion activity in a solution. The standard unit of measurement is the pH unit. A pH of 7 is neutral, less than 7 is acidic, and greater than 7 is basic.

In medical terms, hydrogen-ion concentration is important for maintaining homeostasis within the body. For example, in the stomach, a high hydrogen-ion concentration (low pH) is necessary for the digestion of food. However, in other parts of the body such as blood, a high hydrogen-ion concentration can be harmful and lead to acidosis. Conversely, a low hydrogen-ion concentration (high pH) in the blood can lead to alkalosis. Both acidosis and alkalosis can have serious consequences on various organ systems if not corrected.

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.

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.

Complementary therapies refer to a group of diverse medical and health care systems, practices, and products that are not presently considered an integral part of conventional allopathic medicine. They are often used in conjunction with conventional treatments and are intended to facilitate the physical and emotional well-being of the patient. Complementary therapies can include a wide range of interventions such as acupuncture, chiropractic care, massage therapy, herbal medicine, yoga, meditation, guided imagery, hypnotherapy, and homeopathy, among others. It is important to note that while some complementary therapies have been shown to be effective for certain conditions, others lack scientific evidence of their safety and efficacy. Therefore, it is always recommended to consult with a healthcare provider before starting any new complementary therapy.

Mitochondrial proteins are any proteins that are encoded by the nuclear genome or mitochondrial genome and are located within the mitochondria, an organelle found in eukaryotic cells. These proteins play crucial roles in various cellular processes including energy production, metabolism of lipids, amino acids, and steroids, regulation of calcium homeostasis, and programmed cell death or apoptosis.

Mitochondrial proteins can be classified into two main categories based on their origin:

1. Nuclear-encoded mitochondrial proteins (NEMPs): These are proteins that are encoded by genes located in the nucleus, synthesized in the cytoplasm, and then imported into the mitochondria through specific import pathways. NEMPs make up about 99% of all mitochondrial proteins and are involved in various functions such as oxidative phosphorylation, tricarboxylic acid (TCA) cycle, fatty acid oxidation, and mitochondrial dynamics.

2. Mitochondrial DNA-encoded proteins (MEPs): These are proteins that are encoded by the mitochondrial genome, synthesized within the mitochondria, and play essential roles in the electron transport chain (ETC), a key component of oxidative phosphorylation. The human mitochondrial genome encodes only 13 proteins, all of which are subunits of complexes I, III, IV, and V of the ETC.

Defects in mitochondrial proteins can lead to various mitochondrial disorders, which often manifest as neurological, muscular, or metabolic symptoms due to impaired energy production. These disorders are usually caused by mutations in either nuclear or mitochondrial genes that encode mitochondrial proteins.

ADAM (A Disintegrin And Metalloprotease) proteins are a family of type I transmembrane proteins that contain several distinct domains, including a prodomain, a metalloprotease domain, a disintegrin-like domain, a cysteine-rich domain, a transmembrane domain, and a cytoplasmic tail. These proteins are involved in various biological processes such as cell adhesion, migration, proteolysis, and signal transduction.

ADAM proteins have been found to play important roles in many physiological and pathological conditions, including fertilization, neurodevelopment, inflammation, and cancer metastasis. For example, ADAM12 is involved in the fusion of myoblasts during muscle development, while ADAM17 (also known as TACE) plays a crucial role in the shedding of membrane-bound proteins such as tumor necrosis factor-alpha and epidermal growth factor receptor ligands.

Abnormalities in ADAM protein function have been implicated in various diseases, including cancer, Alzheimer's disease, and arthritis. Therefore, understanding the structure and function of these proteins has important implications for the development of novel therapeutic strategies.

An erythrocyte, also known as a red blood cell, is a type of cell that circulates in the blood and is responsible for transporting oxygen throughout the body. The erythrocyte membrane refers to the thin, flexible barrier that surrounds the erythrocyte and helps to maintain its shape and stability.

The erythrocyte membrane is composed of a lipid bilayer, which contains various proteins and carbohydrates. These components help to regulate the movement of molecules into and out of the erythrocyte, as well as provide structural support and protection for the cell.

The main lipids found in the erythrocyte membrane are phospholipids and cholesterol, which are arranged in a bilayer structure with the hydrophilic (water-loving) heads facing outward and the hydrophobic (water-fearing) tails facing inward. This arrangement helps to maintain the integrity of the membrane and prevent the leakage of cellular components.

The proteins found in the erythrocyte membrane include integral proteins, which span the entire width of the membrane, and peripheral proteins, which are attached to the inner or outer surface of the membrane. These proteins play a variety of roles, such as transporting molecules across the membrane, maintaining the shape of the erythrocyte, and interacting with other cells and proteins in the body.

The carbohydrates found in the erythrocyte membrane are attached to the outer surface of the membrane and help to identify the cell as part of the body's own immune system. They also play a role in cell-cell recognition and adhesion.

Overall, the erythrocyte membrane is a complex and dynamic structure that plays a critical role in maintaining the function and integrity of red blood cells.

Protein isoforms are different forms or variants of a protein that are produced from a single gene through the process of alternative splicing, where different exons (or parts of exons) are included in the mature mRNA molecule. This results in the production of multiple, slightly different proteins that share a common core structure but have distinct sequences and functions. Protein isoforms can also arise from genetic variations such as single nucleotide polymorphisms or mutations that alter the protein-coding sequence of a gene. These differences in protein sequence can affect the stability, localization, activity, or interaction partners of the protein isoform, leading to functional diversity and specialization within cells and organisms.

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.

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

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

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

A plasmid is a small, circular, double-stranded DNA molecule that is separate from the chromosomal DNA of a bacterium or other organism. Plasmids are typically not essential for the survival of the organism, but they can confer beneficial traits such as antibiotic resistance or the ability to degrade certain types of pollutants.

Plasmids are capable of replicating independently of the chromosomal DNA and can be transferred between bacteria through a process called conjugation. They often contain genes that provide resistance to antibiotics, heavy metals, and other environmental stressors. Plasmids have also been engineered for use in molecular biology as cloning vectors, allowing scientists to replicate and manipulate specific DNA sequences.

Plasmids are important tools in genetic engineering and biotechnology because they can be easily manipulated and transferred between organisms. They have been used to produce vaccines, diagnostic tests, and genetically modified organisms (GMOs) for various applications, including agriculture, medicine, and industry.

Temperature, in a medical context, is a measure of the degree of hotness or coldness of a body or environment. It is usually measured using a thermometer and reported in degrees Celsius (°C), degrees Fahrenheit (°F), or kelvin (K). In the human body, normal core temperature ranges from about 36.5-37.5°C (97.7-99.5°F) when measured rectally, and can vary slightly depending on factors such as time of day, physical activity, and menstrual cycle. Elevated body temperature is a common sign of infection or inflammation, while abnormally low body temperature can indicate hypothermia or other medical conditions.

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.

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.

Genetic models are theoretical frameworks used in genetics to describe and explain the inheritance patterns and genetic architecture of traits, diseases, or phenomena. These models are based on mathematical equations and statistical methods that incorporate information about gene frequencies, modes of inheritance, and the effects of environmental factors. They can be used to predict the probability of certain genetic outcomes, to understand the genetic basis of complex traits, and to inform medical management and treatment decisions.

There are several types of genetic models, including:

1. Mendelian models: These models describe the inheritance patterns of simple genetic traits that follow Mendel's laws of segregation and independent assortment. Examples include autosomal dominant, autosomal recessive, and X-linked inheritance.
2. Complex trait models: These models describe the inheritance patterns of complex traits that are influenced by multiple genes and environmental factors. Examples include heart disease, diabetes, and cancer.
3. Population genetics models: These models describe the distribution and frequency of genetic variants within populations over time. They can be used to study evolutionary processes, such as natural selection and genetic drift.
4. Quantitative genetics models: These models describe the relationship between genetic variation and phenotypic variation in continuous traits, such as height or IQ. They can be used to estimate heritability and to identify quantitative trait loci (QTLs) that contribute to trait variation.
5. Statistical genetics models: These models use statistical methods to analyze genetic data and infer the presence of genetic associations or linkage. They can be used to identify genetic risk factors for diseases or traits.

Overall, genetic models are essential tools in genetics research and medical genetics, as they allow researchers to make predictions about genetic outcomes, test hypotheses about the genetic basis of traits and diseases, and develop strategies for prevention, diagnosis, and treatment.

Catalysis is the process of increasing the rate of a chemical reaction by adding a substance known as a catalyst, which remains unchanged at the end of the reaction. A catalyst lowers the activation energy required for the reaction to occur, thereby allowing the reaction to proceed more quickly and efficiently. This can be particularly important in biological systems, where enzymes act as catalysts to speed up metabolic reactions that are essential for life.

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.

Proteolysis is the biological process of breaking down proteins into smaller polypeptides or individual amino acids by the action of enzymes called proteases. This process is essential for various physiological functions, including digestion, protein catabolism, cell signaling, and regulation of numerous biological activities. Dysregulation of proteolysis can contribute to several pathological conditions, such as cancer, neurodegenerative diseases, and inflammatory disorders.

Glycosylation is the enzymatic process of adding a sugar group, or glycan, to a protein, lipid, or other organic molecule. This post-translational modification plays a crucial role in modulating various biological functions, such as protein stability, trafficking, and ligand binding. The structure and composition of the attached glycans can significantly influence the functional properties of the modified molecule, contributing to cell-cell recognition, signal transduction, and immune response regulation. Abnormal glycosylation patterns have been implicated in several disease states, including cancer, diabetes, and neurodegenerative disorders.

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.

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.

Oxyntomodulin is a hormone that is produced and released by the intestines in response to food consumption. It is a 37-amino acid peptide, derived from the preproglucagon gene, which also encodes several other related peptides such as glucagon and GLP-1 (Glucagon-Like Peptide-1).

Oxyntomodulin has two primary effects on the body:

1. Incretin effect: Oxyntomodulin acts as an incretin hormone, which means that it enhances the secretion of insulin and inhibits the release of glucagon in response to a meal. This helps to regulate blood sugar levels and maintain metabolic homeostasis.
2. Appetite regulation: Oxyntomodulin also acts on the hypothalamus to suppress appetite and promote weight loss. It does this by activating receptors in the brain that signal satiety, leading to a decrease in food intake.

Overall, oxyntomodulin plays an important role in regulating energy balance, glucose metabolism, and body weight.

Steroid hydroxylases are enzymes that catalyze the addition of a hydroxyl group (-OH) to a steroid molecule. These enzymes are located in the endoplasmic reticulum and play a crucial role in the biosynthesis of various steroid hormones, such as cortisol, aldosterone, and sex hormones. The hydroxylation reaction catalyzed by these enzymes increases the polarity and solubility of steroids, allowing them to be further metabolized and excreted from the body.

The most well-known steroid hydroxylases are part of the cytochrome P450 family, specifically CYP11A1, CYP11B1, CYP11B2, CYP17A1, CYP19A1, and CYP21A2. Each enzyme has a specific function in steroid biosynthesis, such as converting cholesterol to pregnenolone (CYP11A1), hydroxylating the 11-beta position of steroids (CYP11B1 and CYP11B2), or performing multiple hydroxylation reactions in the synthesis of sex hormones (CYP17A1, CYP19A1, and CYP21A2).

Defects in these enzymes can lead to various genetic disorders, such as congenital adrenal hyperplasia, which is characterized by impaired steroid hormone biosynthesis.

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.

Trypsin is a proteolytic enzyme, specifically a serine protease, that is secreted by the pancreas as an inactive precursor, trypsinogen. Trypsinogen is converted into its active form, trypsin, in the small intestine by enterokinase, which is produced by the intestinal mucosa.

Trypsin plays a crucial role in digestion by cleaving proteins into smaller peptides at specific arginine and lysine residues. This enzyme helps to break down dietary proteins into amino acids, allowing for their absorption and utilization by the body. Additionally, trypsin can activate other zymogenic pancreatic enzymes, such as chymotrypsinogen and procarboxypeptidases, thereby contributing to overall protein digestion.

High-performance liquid chromatography (HPLC) is a type of chromatography that separates and analyzes compounds based on their interactions with a stationary phase and a mobile phase under high pressure. The mobile phase, which can be a gas or liquid, carries the sample mixture through a column containing the stationary phase.

In HPLC, the mobile phase is a liquid, and it is pumped through the column at high pressures (up to several hundred atmospheres) to achieve faster separation times and better resolution than other types of liquid chromatography. The stationary phase can be a solid or a liquid supported on a solid, and it interacts differently with each component in the sample mixture, causing them to separate as they travel through the column.

HPLC is widely used in analytical chemistry, pharmaceuticals, biotechnology, and other fields to separate, identify, and quantify compounds present in complex mixtures. It can be used to analyze a wide range of substances, including drugs, hormones, vitamins, pigments, flavors, and pollutants. HPLC is also used in the preparation of pure samples for further study or use.

Saccharomyces cerevisiae proteins are the proteins that are produced by the budding yeast, Saccharomyces cerevisiae. This organism is a single-celled eukaryote that has been widely used as a model organism in scientific research for many years due to its relatively simple genetic makeup and its similarity to higher eukaryotic cells.

The genome of Saccharomyces cerevisiae has been fully sequenced, and it is estimated to contain approximately 6,000 genes that encode proteins. These proteins play a wide variety of roles in the cell, including catalyzing metabolic reactions, regulating gene expression, maintaining the structure of the cell, and responding to environmental stimuli.

Many Saccharomyces cerevisiae proteins have human homologs and are involved in similar biological processes, making this organism a valuable tool for studying human disease. For example, many of the proteins involved in DNA replication, repair, and recombination in yeast have human counterparts that are associated with cancer and other diseases. By studying these proteins in yeast, researchers can gain insights into their function and regulation in humans, which may lead to new treatments for disease.

Inulin is a soluble fiber that is not digestible by human enzymes. It is a fructan, a type of carbohydrate made up of chains of fructose molecules, and is found in various plants such as chicory root, Jerusalem artichokes, and onions.

Inulin has a number of potential health benefits, including promoting the growth of beneficial bacteria in the gut (prebiotic effect), slowing down the absorption of sugar to help regulate blood glucose levels, and increasing feelings of fullness to aid in weight management. It is often used as a functional food ingredient or dietary supplement for these purposes.

Inulin can also be used as a diagnostic tool in medical testing to measure kidney function, as it is excreted unchanged in the urine.

"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.

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.

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.

Alpha 1-antitrypsin (AAT, or α1-antiproteinase, A1AP) is a protein that is primarily produced by the liver and released into the bloodstream. It belongs to a group of proteins called serine protease inhibitors, which help regulate inflammation and protect tissues from damage caused by enzymes involved in the immune response.

Alpha 1-antitrypsin is particularly important for protecting the lungs from damage caused by neutrophil elastase, an enzyme released by white blood cells called neutrophils during inflammation. In the lungs, AAT binds to and inhibits neutrophil elastase, preventing it from degrading the extracellular matrix and damaging lung tissue.

Deficiency in alpha 1-antitrypsin can lead to chronic obstructive pulmonary disease (COPD) and liver disease. The most common cause of AAT deficiency is a genetic mutation that results in abnormal folding and accumulation of the protein within liver cells, leading to reduced levels of functional AAT in the bloodstream. This condition is called alpha 1-antitrypsin deficiency (AATD) and can be inherited in an autosomal codominant manner. Individuals with severe AATD may require augmentation therapy with intravenous infusions of purified human AAT to help prevent lung damage.

Protease inhibitors are a class of antiviral drugs that are used to treat infections caused by retroviruses, such as the human immunodeficiency virus (HIV), which is responsible for causing AIDS. These drugs work by blocking the activity of protease enzymes, which are necessary for the replication and multiplication of the virus within infected cells.

Protease enzymes play a crucial role in the life cycle of retroviruses by cleaving viral polyproteins into functional units that are required for the assembly of new viral particles. By inhibiting the activity of these enzymes, protease inhibitors prevent the virus from replicating and spreading to other cells, thereby slowing down the progression of the infection.

Protease inhibitors are often used in combination with other antiretroviral drugs as part of highly active antiretroviral therapy (HAART) for the treatment of HIV/AIDS. Common examples of protease inhibitors include saquinavir, ritonavir, indinavir, and atazanavir. While these drugs have been successful in improving the outcomes of people living with HIV/AIDS, they can also cause side effects such as nausea, diarrhea, headaches, and lipodystrophy (changes in body fat distribution).

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

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

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

In a medical context, "hot temperature" is not a standard medical term with a specific definition. However, it is often used in relation to fever, which is a common symptom of illness. A fever is typically defined as a body temperature that is higher than normal, usually above 38°C (100.4°F) for adults and above 37.5-38°C (99.5-101.3°F) for children, depending on the source.

Therefore, when a medical professional talks about "hot temperature," they may be referring to a body temperature that is higher than normal due to fever or other causes. It's important to note that a high environmental temperature can also contribute to an elevated body temperature, so it's essential to consider both the body temperature and the environmental temperature when assessing a patient's condition.

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

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

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

Metabolic networks and pathways refer to the complex interconnected series of biochemical reactions that occur within cells to maintain life. These reactions are catalyzed by enzymes and are responsible for the conversion of nutrients into energy, as well as the synthesis and breakdown of various molecules required for cellular function.

A metabolic pathway is a series of chemical reactions that occur in a specific order, with each reaction being catalyzed by a different enzyme. These pathways are often interconnected, forming a larger network of interactions known as a metabolic network.

Metabolic networks can be represented as complex diagrams or models, which show the relationships between different pathways and the flow of matter and energy through the system. These networks can help researchers to understand how cells regulate their metabolism in response to changes in their environment, and how disruptions to these networks can lead to disease.

Some common examples of metabolic pathways include glycolysis, the citric acid cycle (also known as the Krebs cycle), and the pentose phosphate pathway. Each of these pathways plays a critical role in maintaining cellular homeostasis and providing energy for cellular functions.

Gene deletion is a type of mutation where a segment of DNA, containing one or more genes, is permanently lost or removed from a chromosome. This can occur due to various genetic mechanisms such as homologous recombination, non-homologous end joining, or other types of genomic rearrangements.

The deletion of a gene can have varying effects on the organism, depending on the function of the deleted gene and its importance for normal physiological processes. If the deleted gene is essential for survival, the deletion may result in embryonic lethality or developmental abnormalities. However, if the gene is non-essential or has redundant functions, the deletion may not have any noticeable effects on the organism's phenotype.

Gene deletions can also be used as a tool in genetic research to study the function of specific genes and their role in various biological processes. For example, researchers may use gene deletion techniques to create genetically modified animal models to investigate the impact of gene deletion on disease progression or development.

Staphylococcal Protein A (SpA) is a cell wall-associated protein found on many strains of the bacterium Staphylococcus aureus. It plays an important role in the pathogenesis of staphylococcal infections. SpA has several domains that allow it to bind to various host proteins, including immunoglobulins (Igs), complement components, and fibrinogen.

The protein A's ability to bind to the Fc region of Igs, particularly IgG, enables it to inhibit phagocytosis by masking the antibodies' binding sites, thus helping the bacterium evade the host immune system. Additionally, SpA can activate complement component C1 and initiate the classical complement pathway, leading to the release of anaphylatoxins and the formation of the membrane attack complex, which can cause tissue damage.

Furthermore, SpA's binding to fibrinogen promotes bacterial adherence and colonization of host tissues, contributing to the establishment of infection. Overall, Staphylococcal Protein A is a crucial virulence factor in S. aureus infections, making it an important target for the development of novel therapeutic strategies.

Haptoglobins are proteins found in the blood that bind to free hemoglobin, which is released when red blood cells break down. The resulting complex is then removed from the bloodstream by the liver, preventing the loss of iron and potential kidney damage caused by the breakdown products of hemoglobin. Haptoglobins are produced in the liver and their levels can be measured to help diagnose various medical conditions such as hemolytic anemia, liver disease, and inflammation.

Tissue distribution, in the context of pharmacology and toxicology, refers to the way that a drug or xenobiotic (a chemical substance found within an organism that is not naturally produced by or expected to be present within that organism) is distributed throughout the body's tissues after administration. It describes how much of the drug or xenobiotic can be found in various tissues and organs, and is influenced by factors such as blood flow, lipid solubility, protein binding, and the permeability of cell membranes. Understanding tissue distribution is important for predicting the potential effects of a drug or toxin on different parts of the body, and for designing drugs with improved safety and efficacy profiles.

Chymases are a type of enzyme that belong to the family of serine proteases. They are found in various tissues and organs, including the heart, lungs, and immune cells called mast cells. Chymases play a role in several physiological and pathological processes, such as inflammation, tissue remodeling, and blood pressure regulation.

One of the most well-known chymases is found in the mast cells and is often referred to as "mast cell chymase." This enzyme can cleave and activate various proteins, including angiotensin I to angiotensin II, a potent vasoconstrictor that increases blood pressure. Chymases have also been implicated in the development of cardiovascular diseases, such as hypertension and heart failure, as well as respiratory diseases like asthma and chronic obstructive pulmonary disease (COPD).

In summary, chymases are a group of serine protease enzymes that play important roles in various physiological and pathological processes, particularly in inflammation, tissue remodeling, and blood pressure regulation.

"Plant proteins" refer to the proteins that are derived from plant sources. These can include proteins from legumes such as beans, lentils, and peas, as well as proteins from grains like wheat, rice, and corn. Other sources of plant proteins include nuts, seeds, and vegetables.

Plant proteins are made up of individual amino acids, which are the building blocks of protein. While animal-based proteins typically contain all of the essential amino acids that the body needs to function properly, many plant-based proteins may be lacking in one or more of these essential amino acids. However, by consuming a variety of plant-based foods throughout the day, it is possible to get all of the essential amino acids that the body needs from plant sources alone.

Plant proteins are often lower in calories and saturated fat than animal proteins, making them a popular choice for those following a vegetarian or vegan diet, as well as those looking to maintain a healthy weight or reduce their risk of chronic diseases such as heart disease and cancer. Additionally, plant proteins have been shown to have a number of health benefits, including improving gut health, reducing inflammation, and supporting muscle growth and repair.

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.

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.

Matrix-Assisted Laser Desorption/Ionization Mass Spectrometry (MALDI-MS) is a type of mass spectrometry that is used to analyze large biomolecules such as proteins and peptides. In this technique, the sample is mixed with a matrix compound, which absorbs laser energy and helps to vaporize and ionize the analyte molecules.

The matrix-analyte mixture is then placed on a target plate and hit with a laser beam, causing the matrix and analyte molecules to desorb from the plate and become ionized. The ions are then accelerated through an electric field and into a mass analyzer, which separates them based on their mass-to-charge ratio.

The separated ions are then detected and recorded as a mass spectrum, which can be used to identify and quantify the analyte molecules present in the sample. MALDI-MS is particularly useful for the analysis of complex biological samples, such as tissue extracts or biological fluids, because it allows for the detection and identification of individual components within those mixtures.

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.

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.

I'm sorry for any confusion, but "Isoflurophate" does not appear to be a recognized term in medical or scientific literature. It is possible that there may be a spelling error or typo in the term you are looking for. If you meant "Isoflurane," which is a commonly used anesthetic in medical and surgical procedures, I can provide a definition for that.

Isoflurane: A volatile halogenated ether liquid used as an inhalational general anesthetic agent. It has a rapid onset and offset of action, making it useful for both induction and maintenance of anesthesia. Isoflurane is also known to have bronchodilatory properties, which can be beneficial in patients with reactive airway disease or asthma.

Cyclic peptides are a type of peptides in which the N-terminus and C-terminus of the peptide chain are linked to form a circular structure. This is in contrast to linear peptides, which have a straight peptide backbone with a free N-terminus and C-terminus. The cyclization of peptides can occur through various mechanisms, including the formation of an amide bond between the N-terminal amino group and the C-terminal carboxylic acid group (head-to-tail cyclization), or through the formation of a bond between side chain functional groups.

Cyclic peptides have unique structural and chemical properties that make them valuable in medical and therapeutic applications. For example, they are more resistant to degradation by enzymes compared to linear peptides, which can increase their stability and half-life in the body. Additionally, the cyclic structure allows for greater conformational rigidity, which can enhance their binding affinity and specificity to target molecules.

Cyclic peptides have been explored as potential therapeutics for a variety of diseases, including cancer, infectious diseases, and neurological disorders. They have also been used as tools in basic research to study protein-protein interactions and cell signaling pathways.

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.

Pentose phosphates are monosaccharides that contain five carbon atoms and one phosphate group. They play a crucial role in various metabolic pathways, including the pentose phosphate pathway (PPP), which is a major source of NADPH and ribose-5-phosphate for the synthesis of nucleotides.

The pentose phosphate pathway involves two main phases: the oxidative phase and the non-oxidative phase. In the oxidative phase, glucose-6-phosphate is converted to ribulose-5-phosphate, producing NADPH and CO2 as byproducts. Ribulose-5-phosphate can then be further metabolized in the non-oxidative phase to produce other pentose phosphates or converted back to glucose-6-phosphate through a series of reactions.

Pentose phosphates are also important intermediates in the synthesis of nucleotides, coenzymes, and other metabolites. Abnormalities in pentose phosphate pathway enzymes can lead to various metabolic disorders, such as defects in erythrocyte function and increased susceptibility to oxidative stress.

An amino acid substitution is a type of mutation in which one amino acid in a protein is replaced by another. This occurs when there is a change in the DNA sequence that codes for a particular amino acid in a protein. The genetic code is redundant, meaning that most amino acids are encoded by more than one codon (a sequence of three nucleotides). As a result, a single base pair change in the DNA sequence may not necessarily lead to an amino acid substitution. However, if a change does occur, it can have a variety of effects on the protein's structure and function, depending on the nature of the substituted amino acids. Some substitutions may be harmless, while others may alter the protein's activity or stability, leading to disease.

Antinuclear antibodies (ANA) are a type of autoantibody that target structures found in the nucleus of a cell. These antibodies are produced by the immune system and attack the body's own cells and tissues, leading to inflammation and damage. The presence of ANA is often used as a marker for certain autoimmune diseases, such as systemic lupus erythematosus (SLE), Sjogren's syndrome, rheumatoid arthritis, scleroderma, and polymyositis.

ANA can be detected through a blood test called the antinuclear antibody test. A positive result indicates the presence of ANA in the blood, but it does not necessarily mean that a person has an autoimmune disease. Further testing is usually needed to confirm a diagnosis and determine the specific type of autoantibodies present.

It's important to note that ANA can also be found in healthy individuals, particularly as they age. Therefore, the test results should be interpreted in conjunction with other clinical findings and symptoms.

Cholestanetriol 26-monooxygenase is an enzyme that is involved in the metabolism of bile acids and steroids in the body. This enzyme is responsible for adding a hydroxyl group (-OH) to the cholestanetriol molecule at position 26, which is a critical step in the conversion of cholestanetriol to bile acids.

The gene that encodes this enzyme is called CYP3A4, which is located on chromosome 7 in humans. Mutations in this gene can lead to various metabolic disorders, including impaired bile acid synthesis and altered steroid hormone metabolism.

Deficiency or dysfunction of cholestanetriol 26-monooxygenase has been associated with several diseases, such as liver disease, cerebrotendinous xanthomatosis, and some forms of cancer. Therefore, understanding the function and regulation of this enzyme is essential for developing new therapies and treatments for these conditions.

Immunodiffusion is a laboratory technique used in immunology to detect and measure the presence of specific antibodies or antigens in a sample. It is based on the principle of diffusion, where molecules move from an area of high concentration to an area of low concentration until they reach equilibrium. In this technique, a sample containing an unknown quantity of antigen or antibody is placed in a gel or agar medium that contains a known quantity of antibody or antigen, respectively.

The two substances then diffuse towards each other and form a visible precipitate at the point where they meet and reach equivalence, which indicates the presence and quantity of the specific antigen or antibody in the sample. There are several types of immunodiffusion techniques, including radial immunodiffusion (RID) and double immunodiffusion (Ouchterlony technique). These techniques are widely used in diagnostic laboratories to identify and measure various antigens and antibodies, such as those found in infectious diseases, autoimmune disorders, and allergic reactions.

Cell respiration is the process by which cells convert biochemical energy from nutrients into adenosine triphosphate (ATP), and then release waste products. The three main stages of cell respiration are glycolysis, the citric acid cycle (also known as the Krebs cycle), and the electron transport chain.

During glycolysis, which takes place in the cytoplasm, glucose is broken down into two molecules of pyruvate, producing a small amount of ATP and reducing power in the form of NADH.

The citric acid cycle occurs in the mitochondria and involves the breakdown of acetyl-CoA (formed from pyruvate) to produce more ATP, NADH, and FADH2.

Finally, the electron transport chain, also located in the mitochondria, uses the energy from NADH and FADH2 to pump protons across the inner mitochondrial membrane, creating a proton gradient. The flow of protons back across the membrane drives the synthesis of ATP, which is used as a source of energy by the cell.

Cell respiration is a crucial process that allows cells to generate the energy they need to perform various functions and maintain homeostasis.

Lupus nephritis is a type of kidney inflammation (nephritis) that can occur in people with systemic lupus erythematosus (SLE), an autoimmune disease. In lupus nephritis, the immune system produces abnormal antibodies that attack the tissues of the kidneys, leading to inflammation and damage. The condition can cause a range of symptoms, including proteinuria (protein in the urine), hematuria (blood in the urine), hypertension (high blood pressure), and eventually kidney failure if left untreated. Lupus nephritis is typically diagnosed through a combination of medical history, physical examination, laboratory tests, and imaging studies. Treatment may include medications to suppress the immune system and control inflammation, such as corticosteroids and immunosuppressive drugs.

Polysaccharides are complex carbohydrates consisting of long chains of monosaccharide units (simple sugars) bonded together by glycosidic linkages. They can be classified based on the type of monosaccharides and the nature of the bonds that connect them.

Polysaccharides have various functions in living organisms. For example, starch and glycogen serve as energy storage molecules in plants and animals, respectively. Cellulose provides structural support in plants, while chitin is a key component of fungal cell walls and arthropod exoskeletons.

Some polysaccharides also have important roles in the human body, such as being part of the extracellular matrix (e.g., hyaluronic acid) or acting as blood group antigens (e.g., ABO blood group substances).

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.

Protein transport, in the context of cellular biology, refers to the process by which proteins are actively moved from one location to another within or between cells. This is a crucial mechanism for maintaining proper cell function and regulation.

Intracellular protein transport involves the movement of proteins within a single cell. Proteins can be transported across membranes (such as the nuclear envelope, endoplasmic reticulum, Golgi apparatus, or plasma membrane) via specialized transport systems like vesicles and transport channels.

Intercellular protein transport refers to the movement of proteins from one cell to another, often facilitated by exocytosis (release of proteins in vesicles) and endocytosis (uptake of extracellular substances via membrane-bound vesicles). This is essential for communication between cells, immune response, and other physiological processes.

It's important to note that any disruption in protein transport can lead to various diseases, including neurological disorders, cancer, and metabolic conditions.

Epithelial cells are types of cells that cover the outer surfaces of the body, line the inner surfaces of organs and glands, and form the lining of blood vessels and body cavities. They provide a protective barrier against the external environment, regulate the movement of materials between the internal and external environments, and are involved in the sense of touch, temperature, and pain. Epithelial cells can be squamous (flat and thin), cuboidal (square-shaped and of equal height), or columnar (tall and narrow) in shape and are classified based on their location and function.

Methylamines are organic compounds that contain a methyl group (CH3) and an amino group (-NH2). They have the general formula of CH3-NH-R, where R can be a hydrogen atom or any organic group. Methylamines are derivatives of ammonia (NH3), in which one or more hydrogen atoms have been replaced by methyl groups.

There are several types of methylamines, including:

1. Methylamine (CH3-NH2): This is the simplest methylamine and is a colorless gas at room temperature with a strong odor. It is highly flammable and reactive.
2. Dimethylamine (CH3)2-NH: This is a colorless liquid at room temperature with an unpleasant fishy odor. It is less reactive than methylamine but still highly flammable.
3. Trimethylamine (CH3)3-N: This is a colorless liquid at room temperature that has a strong, unpleasant odor often described as "fishy." It is less reactive than dimethylamine and is used in various industrial applications.

Methylamines are used in the production of various chemicals, including pesticides, dyes, and pharmaceuticals. They can also be found naturally in some foods and are produced by certain types of bacteria in the body. Exposure to high levels of methylamines can cause irritation to the eyes, skin, and respiratory tract, and prolonged exposure can lead to more serious health effects.

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.

Cosmids are a type of cloning vector, which are self-replicating DNA molecules that can be used to introduce foreign DNA fragments into a host organism. Cosmids are plasmids that contain the cos site from bacteriophage λ, allowing them to be packaged into bacteriophage heads during an in vitro packaging reaction. This enables the transfer of large DNA fragments (up to 45 kb) into a host cell through transduction. Cosmids are widely used in molecular biology for the construction and analysis of genomic libraries, physical mapping, and DNA sequencing.

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.

A homozygote is an individual who has inherited the same allele (version of a gene) from both parents and therefore possesses two identical copies of that allele at a specific genetic locus. This can result in either having two dominant alleles (homozygous dominant) or two recessive alleles (homozygous recessive). In contrast, a heterozygote has inherited different alleles from each parent for a particular gene.

The term "homozygote" is used in genetics to describe the genetic makeup of an individual at a specific locus on their chromosomes. Homozygosity can play a significant role in determining an individual's phenotype (observable traits), as having two identical alleles can strengthen the expression of certain characteristics compared to having just one dominant and one recessive allele.

Gene dosage, in genetic terms, refers to the number of copies of a particular gene present in an organism's genome. Each gene usually has two copies (alleles) in diploid organisms, one inherited from each parent. An increase or decrease in the number of copies of a specific gene can lead to changes in the amount of protein it encodes, which can subsequently affect various biological processes and phenotypic traits.

For example, gene dosage imbalances have been associated with several genetic disorders, such as Down syndrome (trisomy 21), where an individual has three copies of chromosome 21 instead of the typical two copies, leading to developmental delays and intellectual disabilities. Similarly, in certain cases of cancer, gene amplification (an increase in the number of copies of a particular gene) can result in overexpression of oncogenes, contributing to tumor growth and progression.

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.

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

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

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

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

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

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

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.

A haplotype is a group of genes or DNA sequences that are inherited together from a single parent. It refers to a combination of alleles (variant forms of a gene) that are located on the same chromosome and are usually transmitted as a unit. Haplotypes can be useful in tracing genetic ancestry, understanding the genetic basis of diseases, and developing personalized medical treatments.

In population genetics, haplotypes are often used to study patterns of genetic variation within and between populations. By comparing haplotype frequencies across populations, researchers can infer historical events such as migrations, population expansions, and bottlenecks. Additionally, haplotypes can provide information about the evolutionary history of genes and genomic regions.

In clinical genetics, haplotypes can be used to identify genetic risk factors for diseases or to predict an individual's response to certain medications. For example, specific haplotypes in the HLA gene region have been associated with increased susceptibility to certain autoimmune diseases, while other haplotypes in the CYP450 gene family can affect how individuals metabolize drugs.

Overall, haplotypes provide a powerful tool for understanding the genetic basis of complex traits and diseases, as well as for developing personalized medical treatments based on an individual's genetic makeup.

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).

Proinsulin is the precursor protein to insulin, produced in the beta cells of the pancreas. It has a molecular weight of around 9,000 daltons and is composed of three distinct regions: the A-chain, the B-chain, and the C-peptide. The A-chain and B-chain are linked together by disulfide bonds and will eventually become the insulin molecule after a series of enzymatic cleavages. The C-peptide is removed during this process and is released into the bloodstream in equimolar amounts to insulin. Proinsulin levels can be measured in the blood and are sometimes used as a marker for beta cell function in certain clinical settings, such as diagnosing or monitoring insulinoma (a tumor of the pancreas that produces insulin) or assessing the risk of diabetes-related complications.

Immunoblotting, also known as western blotting, is a laboratory technique used in molecular biology and immunogenetics to detect and quantify specific proteins in a complex mixture. This technique combines the electrophoretic separation of proteins by gel electrophoresis with their detection using antibodies that recognize specific epitopes (protein fragments) on the target protein.

The process involves several steps: first, the protein sample is separated based on size through sodium dodecyl sulfate-polyacrylamide gel electrophoresis (SDS-PAGE). Next, the separated proteins are transferred onto a nitrocellulose or polyvinylidene fluoride (PVDF) membrane using an electric field. The membrane is then blocked with a blocking agent to prevent non-specific binding of antibodies.

After blocking, the membrane is incubated with a primary antibody that specifically recognizes the target protein. Following this, the membrane is washed to remove unbound primary antibodies and then incubated with a secondary antibody conjugated to an enzyme such as horseradish peroxidase (HRP) or alkaline phosphatase (AP). The enzyme catalyzes a colorimetric or chemiluminescent reaction that allows for the detection of the target protein.

Immunoblotting is widely used in research and clinical settings to study protein expression, post-translational modifications, protein-protein interactions, and disease biomarkers. It provides high specificity and sensitivity, making it a valuable tool for identifying and quantifying proteins in various biological samples.

Meningococcal infections are caused by the bacterium Neisseria meningitidis, also known as meningococcus. These infections can take several forms, but the most common are meningitis (inflammation of the membranes surrounding the brain and spinal cord) and septicemia (bloodstream infection). Meningococcal infections are contagious and can spread through respiratory droplets or close contact with an infected person. They can be serious and potentially life-threatening, requiring prompt medical attention and treatment with antibiotics. Symptoms of meningococcal meningitis may include fever, headache, stiff neck, and sensitivity to light, while symptoms of septicemia may include fever, chills, rash, and severe muscle pain. Vaccination is available to prevent certain strains of meningococcal disease.

Amino acid repetitive sequences refer to patterns of amino acids that are repeated in a polypeptide chain. These repetitions can vary in length and can be composed of a single type of amino acid or a combination of different types. In some cases, expansions of these repetitive sequences can lead to the production of abnormal proteins that are associated with certain genetic disorders. The expansion of trinucleotide repeats that code for particular amino acids is one example of this phenomenon. These expansions can result in protein misfolding and aggregation, leading to neurodegenerative diseases such as Huntington's disease and spinocerebellar ataxias.

Neisseria meningitidis, Serogroup W-135 is a subtype of the bacterium Neisseria meningitidis, also known as meningococcus. This gram-negative diplococcus is a leading cause of bacterial meningitis and sepsis worldwide. The serogroups of N. meningitidis are defined based on the chemical structure of their capsular polysaccharides, which are essential virulence factors.

Serogroup W-135 is one of the six primary serogroups (A, B, C, W, X, and Y) that account for nearly all meningococcal disease cases globally. The W-135 serogroup has been associated with several outbreaks and sporadic cases of meningitis and sepsis, particularly in the African "meningitis belt," which stretches across the continent from Senegal to Ethiopia. However, it can also cause disease in other parts of the world, including Europe, America, and Asia.

The W-135 serogroup has been a concern due to its association with travel and pilgrimages, such as the Hajj in Saudi Arabia. The Hajj-associated meningococcal disease outbreaks led to the introduction of vaccination requirements for international travelers attending the pilgrimage.

Vaccines are available to protect against N. meningitidis Serogroup W-135, and they are often combined with other serogroups (e.g., MenACWY or MenQuad) to provide broader protection against multiple serogroups. These vaccines have been instrumental in controlling outbreaks and reducing the overall burden of meningococcal disease worldwide.

Phylogeny is the evolutionary history and relationship among biological entities, such as species or genes, based on their shared characteristics. In other words, it refers to the branching pattern of evolution that shows how various organisms have descended from a common ancestor over time. Phylogenetic analysis involves constructing a tree-like diagram called a phylogenetic tree, which depicts the inferred evolutionary relationships among organisms or genes based on molecular sequence data or other types of characters. This information is crucial for understanding the diversity and distribution of life on Earth, as well as for studying the emergence and spread of diseases.

LDL, or low-density lipoprotein, is often referred to as "bad" cholesterol. It is one of the lipoproteins that helps carry cholesterol throughout your body. High levels of LDL cholesterol can lead to a buildup of cholesterol in your arteries, which can increase the risk of heart disease and stroke.

Cholesterol is a type of fat (lipid) that is found in the cells of your body. Your body needs some cholesterol to function properly, but having too much can lead to health problems. LDL cholesterol is one of the two main types of cholesterol; the other is high-density lipoprotein (HDL), or "good" cholesterol.

It's important to keep your LDL cholesterol levels in a healthy range to reduce your risk of developing heart disease and stroke. A healthcare professional can help you determine what your target LDL cholesterol level should be based on your individual health status and risk factors.

Restriction mapping is a technique used in molecular biology to identify the location and arrangement of specific restriction endonuclease recognition sites within a DNA molecule. Restriction endonucleases are enzymes that cut double-stranded DNA at specific sequences, producing fragments of various lengths. By digesting the DNA with different combinations of these enzymes and analyzing the resulting fragment sizes through techniques such as agarose gel electrophoresis, researchers can generate a restriction map - a visual representation of the locations and distances between recognition sites on the DNA molecule. This information is crucial for various applications, including cloning, genome analysis, and genetic engineering.

Isoenzymes, also known as isoforms, are multiple forms of an enzyme that catalyze the same chemical reaction but differ in their amino acid sequence, structure, and/or kinetic properties. They are encoded by different genes or alternative splicing of the same gene. Isoenzymes can be found in various tissues and organs, and they play a crucial role in biological processes such as metabolism, detoxification, and cell signaling. Measurement of isoenzyme levels in body fluids (such as blood) can provide valuable diagnostic information for certain medical conditions, including tissue damage, inflammation, and various diseases.

Cystathionine is a non-proteinogenic amino acid, which means that it is not used in the synthesis of proteins. It is an intermediate in the biosynthetic pathway that converts the amino acid methionine to cysteine in the body. This process involves the removal of a sulfur atom from methionine, resulting in the formation of cystathionine. Further breakdown of cystathionine leads to the production of cysteine and another amino acid called alpha-ketobutyrate.

Cystathionine plays a crucial role in the metabolism of certain sulfur-containing amino acids, and its levels are regulated by an enzyme called cystathionine beta-synthase (CBS). Genetic defects or deficiencies in this enzyme can result in a disorder known as homocystinuria, which is characterized by the accumulation of homocysteine and methionine in the body and an increased risk of various health complications.

In summary, cystathionine is a biologically important amino acid that functions as an intermediate in the conversion of methionine to cysteine, and its levels are tightly regulated by enzymatic processes in the body.

Enzyme stability refers to the ability of an enzyme to maintain its structure and function under various environmental conditions, such as temperature, pH, and the presence of denaturants or inhibitors. A stable enzyme retains its activity and conformation over time and across a range of conditions, making it more suitable for industrial and therapeutic applications.

Enzymes can be stabilized through various methods, including chemical modification, immobilization, and protein engineering. Understanding the factors that affect enzyme stability is crucial for optimizing their use in biotechnology, medicine, and research.

Bacterial outer membrane proteins (OMPs) are a type of protein found in the outer membrane of gram-negative bacteria. The outer membrane is a unique characteristic of gram-negative bacteria, and it serves as a barrier that helps protect the bacterium from hostile environments. OMPs play a crucial role in maintaining the structural integrity and selective permeability of the outer membrane. They are involved in various functions such as nutrient uptake, transport, adhesion, and virulence factor secretion.

OMPs are typically composed of beta-barrel structures that span the bacterial outer membrane. These proteins can be classified into several groups based on their size, function, and structure. Some of the well-known OMP families include porins, autotransporters, and two-partner secretion systems.

Porins are the most abundant type of OMPs and form water-filled channels that allow the passive diffusion of small molecules, ions, and nutrients across the outer membrane. Autotransporters are a diverse group of OMPs that play a role in bacterial pathogenesis by secreting virulence factors or acting as adhesins. Two-partner secretion systems involve the cooperation between two proteins to transport effector molecules across the outer membrane.

Understanding the structure and function of bacterial OMPs is essential for developing new antibiotics and therapies that target gram-negative bacteria, which are often resistant to conventional treatments.

Congenital Adrenal Hyperplasia (CAH) is a group of inherited genetic disorders that affect the adrenal glands, which are triangular-shaped glands located on top of the kidneys. The adrenal glands are responsible for producing several essential hormones, including cortisol, aldosterone, and androgens.

CAH is caused by mutations in genes that code for enzymes involved in the synthesis of these hormones. The most common form of CAH is 21-hydroxylase deficiency, which affects approximately 90% to 95% of all cases. Other less common forms of CAH include 11-beta-hydroxylase deficiency and 3-beta-hydroxysteroid dehydrogenase deficiency.

The severity of the disorder can vary widely, depending on the degree of enzyme deficiency. In severe cases, the lack of cortisol production can lead to life-threatening salt wasting and electrolyte imbalances in newborns. The excess androgens produced due to the enzyme deficiency can also cause virilization, or masculinization, of female fetuses, leading to ambiguous genitalia at birth.

In milder forms of CAH, symptoms may not appear until later in childhood or even adulthood. These may include early puberty, rapid growth followed by premature fusion of the growth plates and short stature, acne, excessive hair growth, irregular menstrual periods, and infertility.

Treatment for CAH typically involves replacing the missing hormones with medications such as hydrocortisone, fludrocortisone, and/or sex hormones. Regular monitoring of hormone levels and careful management of medication doses is essential to prevent complications such as adrenal crisis, growth suppression, and osteoporosis.

In severe cases of CAH, early diagnosis and treatment can help prevent or minimize the risk of serious health problems and improve quality of life. Genetic counseling may also be recommended for affected individuals and their families to discuss the risks of passing on the disorder to future generations.

Bacterial polysaccharides are complex carbohydrates that consist of long chains of sugar molecules (monosaccharides) linked together by glycosidic bonds. They are produced and used by bacteria for various purposes such as:

1. Structural components: Bacterial polysaccharides, such as peptidoglycan and lipopolysaccharide (LPS), play a crucial role in maintaining the structural integrity of bacterial cells. Peptidoglycan is a major component of the bacterial cell wall, while LPS forms the outer layer of the outer membrane in gram-negative bacteria.
2. Nutrient storage: Some bacteria synthesize and store polysaccharides as an energy reserve, similar to how plants store starch. These polysaccharides can be broken down and utilized by the bacterium when needed.
3. Virulence factors: Bacterial polysaccharides can also function as virulence factors, contributing to the pathogenesis of bacterial infections. For example, certain bacteria produce capsular polysaccharides (CPS) that surround and protect the bacterial cells from host immune defenses, allowing them to evade phagocytosis and persist within the host.
4. Adhesins: Some polysaccharides act as adhesins, facilitating the attachment of bacteria to surfaces or host cells. This is important for biofilm formation, which helps bacteria resist environmental stresses and antibiotic treatments.
5. Antigenic properties: Bacterial polysaccharides can be highly antigenic, eliciting an immune response in the host. The antigenicity of these molecules can vary between different bacterial species or even strains within a species, making them useful as targets for vaccines and diagnostic tests.

In summary, bacterial polysaccharides are complex carbohydrates that serve various functions in bacteria, including structural support, nutrient storage, virulence factor production, adhesion, and antigenicity.

Neisseria meningitidis is a Gram-negative, aerobic, bean-shaped diplococcus bacterium. It is one of the leading causes of bacterial meningitis and sepsis (known as meningococcal disease) worldwide. The bacteria can be found in the back of the nose and throat of approximately 10-25% of the general population, particularly in children, teenagers, and young adults, without causing any symptoms or illness. However, when the bacterium invades the bloodstream and spreads to the brain or spinal cord, it can lead to life-threatening infections such as meningitis (inflammation of the membranes surrounding the brain and spinal cord) and septicemia (blood poisoning).

Neisseria meningitidis is classified into 12 serogroups based on the chemical structure of their capsular polysaccharides. The six major serogroups that cause most meningococcal disease worldwide are A, B, C, W, X, and Y. Vaccines are available to protect against some or all of these serogroups.

Meningococcal disease can progress rapidly, leading to severe symptoms such as high fever, headache, stiff neck, confusion, nausea, vomiting, and a rash consisting of purple or red spots. Immediate medical attention is required if someone experiences these symptoms, as meningococcal disease can cause permanent disabilities or death within hours if left untreated.

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 gene is a specific sequence of nucleotides in DNA that carries genetic information. Genes are the fundamental units of heredity and are responsible for the development and function of all living organisms. They code for proteins or RNA molecules, which carry out various functions within cells and are essential for the structure, function, and regulation of the body's tissues and organs.

Each gene has a specific location on a chromosome, and each person inherits two copies of every gene, one from each parent. Variations in the sequence of nucleotides in a gene can lead to differences in traits between individuals, including physical characteristics, susceptibility to disease, and responses to environmental factors.

Medical genetics is the study of genes and their role in health and disease. It involves understanding how genes contribute to the development and progression of various medical conditions, as well as identifying genetic risk factors and developing strategies for prevention, diagnosis, and treatment.

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

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

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

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

Hep G2 cells are a type of human liver cancer cell line that were isolated from a well-differentiated hepatocellular carcinoma (HCC) in a patient with hepatitis C virus (HCV) infection. These cells have the ability to grow and divide indefinitely in culture, making them useful for research purposes. Hep G2 cells express many of the same markers and functions as normal human hepatocytes, including the ability to take up and process lipids and produce bile. They are often used in studies related to hepatitis viruses, liver metabolism, drug toxicity, and cancer biology. It is important to note that Hep G2 cells are tumorigenic and should be handled with care in a laboratory setting.

Meningococcal meningitis is a specific type of bacterial meningitis caused by the bacterium Neisseria meningitidis, also known as meningococcus. Meningitis refers to the inflammation of the meninges, which are the protective membranes covering the brain and spinal cord. When this inflammation is caused by the meningococcal bacteria, it is called meningococcal meningitis.

There are several serogroups of Neisseria meningitidis that can cause invasive disease, with the most common ones being A, B, C, W, and Y. The infection can spread through respiratory droplets or direct contact with an infected person's saliva or secretions, especially when they cough or sneeze.

Meningococcal meningitis is a serious and potentially life-threatening condition that requires immediate medical attention. Symptoms may include sudden onset of fever, severe headache, stiff neck, nausea, vomiting, confusion, and sensitivity to light. In some cases, a rash may also develop, characterized by small purple or red spots that do not blanch when pressed with a glass.

Prevention measures include vaccination against the different serogroups of Neisseria meningitidis, maintaining good personal hygiene, avoiding sharing utensils, cigarettes, or other items that may come into contact with an infected person's saliva, and promptly seeking medical care if symptoms develop.

Serum globulins are a group of proteins present in the liquid portion of blood, known as serum. They are produced by the immune system in response to foreign substances such as bacteria, viruses, and allergens. Serum globulins include several types of immunoglobulins (antibodies), complement components, and other proteins involved in the immune response.

The serum globulin level is often measured as part of a complete blood count (CBC) or a protein electrophoresis test. An elevated serum globulin level may indicate an ongoing infection, inflammation, or an autoimmune disorder. Conversely, a decreased level may suggest a liver or kidney disease, or a malnutrition condition. It is important to note that the interpretation of serum globulin levels should be done in conjunction with other laboratory and clinical findings.

Mitochondria are specialized structures located inside cells that convert the energy from food into ATP (adenosine triphosphate), which is the primary form of energy used by cells. They are often referred to as the "powerhouses" of the cell because they generate most of the cell's supply of chemical energy. Mitochondria are also involved in various other cellular processes, such as signaling, differentiation, and apoptosis (programmed cell death).

Mitochondria have their own DNA, known as mitochondrial DNA (mtDNA), which is inherited maternally. This means that mtDNA is passed down from the mother to her offspring through the egg cells. Mitochondrial dysfunction has been linked to a variety of diseases and conditions, including neurodegenerative disorders, diabetes, and aging.

Lipodystrophy is a medical condition characterized by abnormal distribution or absence of fat (adipose tissue) in the body. It can lead to metabolic complications such as insulin resistance, diabetes mellitus, high levels of fats in the blood (dyslipidemia), and liver disease. There are different types of lipodystrophy, including congenital generalized lipodystrophy, acquired generalized lipodystrophy, and partial lipodystrophy, which can affect different parts of the body and have varying symptoms and causes.

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.

Sterol Regulatory Element Binding Protein 2 (SREBP-2) is a transcription factor that plays a crucial role in the regulation of cholesterol homeostasis in the body. It is a member of the SREBP family, which also includes SREBP-1a and SREBP-1c, and is encoded by the SREBF2 gene.

SREBP-2 is primarily involved in the regulation of genes that are necessary for cholesterol synthesis and uptake. When cholesterol levels in the body are low, SREBP-2 gets activated and moves from the endoplasmic reticulum to the Golgi apparatus, where it undergoes proteolytic cleavage to release its active form. The active SREBP-2 then translocates to the nucleus and binds to sterol regulatory elements (SREs) in the promoter regions of target genes, thereby inducing their transcription.

The target genes of SREBP-2 include HMG-CoA reductase, which is a rate-limiting enzyme in cholesterol synthesis, and LDL receptor, which is responsible for the uptake of low-density lipoprotein (LDL) or "bad" cholesterol from the bloodstream. By upregulating the expression of these genes, SREBP-2 helps to increase cholesterol levels in the body and maintain cholesterol homeostasis.

Dysregulation of SREBP-2 has been implicated in various diseases, including atherosclerosis, cardiovascular disease, and cancer.

X-ray crystallography is a technique used in structural biology to determine the three-dimensional arrangement of atoms in a crystal lattice. In this method, a beam of X-rays is directed at a crystal and diffracts, or spreads out, into a pattern of spots called reflections. The intensity and angle of each reflection are measured and used to create an electron density map, which reveals the position and type of atoms in the crystal. This information can be used to determine the molecular structure of a compound, including its shape, size, and chemical bonds. X-ray crystallography is a powerful tool for understanding the structure and function of biological macromolecules such as proteins and nucleic acids.

Hydrolases are a class of enzymes that help facilitate the breakdown of various types of chemical bonds through a process called hydrolysis, which involves the addition of water. These enzymes catalyze the cleavage of bonds in substrates by adding a molecule of water, leading to the formation of two or more smaller molecules.

Hydrolases play a crucial role in many biological processes, including digestion, metabolism, and detoxification. They can act on a wide range of substrates, such as proteins, lipids, carbohydrates, and nucleic acids, breaking them down into smaller units that can be more easily absorbed or utilized by the body.

Examples of hydrolases include:

1. Proteases: enzymes that break down proteins into smaller peptides or amino acids.
2. Lipases: enzymes that hydrolyze lipids, such as triglycerides, into fatty acids and glycerol.
3. Amylases: enzymes that break down complex carbohydrates, like starches, into simpler sugars, such as glucose.
4. Nucleases: enzymes that cleave nucleic acids, such as DNA or RNA, into smaller nucleotides or oligonucleotides.
5. Phosphatases: enzymes that remove phosphate groups from various substrates, including proteins and lipids.
6. Esterases: enzymes that hydrolyze ester bonds in a variety of substrates, such as those found in some drugs or neurotransmitters.

Hydrolases are essential for maintaining proper cellular function and homeostasis, and their dysregulation can contribute to various diseases and disorders.

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.

Disulfides are a type of organic compound that contains a sulfur-sulfur bond. In the context of biochemistry and medicine, disulfide bonds are often found in proteins, where they play a crucial role in maintaining their three-dimensional structure and function. These bonds form when two sulfhydryl groups (-SH) on cysteine residues within a protein molecule react with each other, releasing a molecule of water and creating a disulfide bond (-S-S-) between the two cysteines. Disulfide bonds can be reduced back to sulfhydryl groups by various reducing agents, which is an important process in many biological reactions. The formation and reduction of disulfide bonds are critical for the proper folding, stability, and activity of many proteins, including those involved in various physiological processes and diseases.

'Cercopithecus aethiops' is the scientific name for the monkey species more commonly known as the green monkey. It belongs to the family Cercopithecidae and is native to western Africa. The green monkey is omnivorous, with a diet that includes fruits, nuts, seeds, insects, and small vertebrates. They are known for their distinctive greenish-brown fur and long tail. Green monkeys are also important animal models in biomedical research due to their susceptibility to certain diseases, such as SIV (simian immunodeficiency virus), which is closely related to HIV.

The Golgi apparatus, also known as the Golgi complex or simply the Golgi, is a membrane-bound organelle found in the cytoplasm of most eukaryotic cells. It plays a crucial role in the processing, sorting, and packaging of proteins and lipids for transport to their final destinations within the cell or for secretion outside the cell.

The Golgi apparatus consists of a series of flattened, disc-shaped sacs called cisternae, which are stacked together in a parallel arrangement. These stacks are often interconnected by tubular structures called tubules or vesicles. The Golgi apparatus has two main faces: the cis face, which is closest to the endoplasmic reticulum (ER) and receives proteins and lipids directly from the ER; and the trans face, which is responsible for sorting and dispatching these molecules to their final destinations.

The Golgi apparatus performs several essential functions in the cell:

1. Protein processing: After proteins are synthesized in the ER, they are transported to the cis face of the Golgi apparatus, where they undergo various post-translational modifications, such as glycosylation (the addition of sugar molecules) and sulfation. These modifications help determine the protein's final structure, function, and targeting.
2. Lipid modification: The Golgi apparatus also modifies lipids by adding or removing different functional groups, which can influence their properties and localization within the cell.
3. Protein sorting and packaging: Once proteins and lipids have been processed, they are sorted and packaged into vesicles at the trans face of the Golgi apparatus. These vesicles then transport their cargo to various destinations, such as lysosomes, plasma membrane, or extracellular space.
4. Intracellular transport: The Golgi apparatus serves as a central hub for intracellular trafficking, coordinating the movement of vesicles and other transport carriers between different organelles and cellular compartments.
5. Cell-cell communication: Some proteins that are processed and packaged in the Golgi apparatus are destined for secretion, playing crucial roles in cell-cell communication and maintaining tissue homeostasis.

In summary, the Golgi apparatus is a vital organelle involved in various cellular processes, including post-translational modification, sorting, packaging, and intracellular transport of proteins and lipids. Its proper functioning is essential for maintaining cellular homeostasis and overall organismal health.

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.

A bacterial gene is a segment of DNA (or RNA in some viruses) that contains the genetic information necessary for the synthesis of a functional bacterial protein or RNA molecule. These genes are responsible for encoding various characteristics and functions of bacteria such as metabolism, reproduction, and resistance to antibiotics. They can be transmitted between bacteria through horizontal gene transfer mechanisms like conjugation, transformation, and transduction. Bacterial genes are often organized into operons, which are clusters of genes that are transcribed together as a single mRNA molecule.

It's important to note that the term "bacterial gene" is used to describe genetic elements found in bacteria, but not all genetic elements in bacteria are considered genes. For example, some DNA sequences may not encode functional products and are therefore not considered genes. Additionally, some bacterial genes may be plasmid-borne or phage-borne, rather than being located on the bacterial chromosome.

Introns are non-coding sequences of DNA that are present within the genes of eukaryotic organisms, including plants, animals, and humans. Introns are removed during the process of RNA splicing, in which the initial RNA transcript is cut and reconnected to form a mature, functional RNA molecule.

After the intron sequences are removed, the remaining coding sequences, known as exons, are joined together to create a continuous stretch of genetic information that can be translated into a protein or used to produce non-coding RNAs with specific functions. The removal of introns allows for greater flexibility in gene expression and regulation, enabling the generation of multiple proteins from a single gene through alternative splicing.

In summary, introns are non-coding DNA sequences within genes that are removed during RNA processing to create functional RNA molecules or proteins.

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.

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

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

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

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.

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.

Molecular evolution is the process of change in the DNA sequence or protein structure over time, driven by mechanisms such as mutation, genetic drift, gene flow, and natural selection. It refers to the evolutionary study of changes in DNA, RNA, and proteins, and how these changes accumulate and lead to new species and diversity of life. Molecular evolution can be used to understand the history and relationships among different organisms, as well as the functional consequences of genetic changes.

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.

"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.

Streptococcus agalactiae, also known as Group B Streptococcus (GBS), is a type of bacteria that commonly colonizes the gastrointestinal and genitourinary tracts of humans. It is Gram-positive, facultatively anaerobic, and forms chains when viewed under the microscope.

While S. agalactiae can be carried asymptomatically by many adults, it can cause serious infections in newborns, pregnant women, elderly individuals, and people with weakened immune systems. In newborns, GBS can lead to sepsis, pneumonia, and meningitis, which can result in long-term health complications or even be fatal if left untreated.

Pregnant women are often screened for GBS colonization during the third trimester of pregnancy, and those who test positive may receive intrapartum antibiotics to reduce the risk of transmission to their newborns during delivery.

Blood is the fluid that circulates in the body of living organisms, carrying oxygen and nutrients to the cells and removing carbon dioxide and other waste products. It is composed of red and white blood cells suspended in a liquid called plasma. The main function of blood is to transport oxygen from the lungs to the body's tissues and carbon dioxide from the tissues to the lungs. It also transports nutrients, hormones, and other substances to the cells and removes waste products from them. Additionally, blood plays a crucial role in the body's immune system by helping to fight infection and disease.

"Competitive binding" is a term used in pharmacology and biochemistry to describe the behavior of two or more molecules (ligands) competing for the same binding site on a target protein or receptor. In this context, "binding" refers to the physical interaction between a ligand and its target.

When a ligand binds to a receptor, it can alter the receptor's function, either activating or inhibiting it. If multiple ligands compete for the same binding site, they will compete to bind to the receptor. The ability of each ligand to bind to the receptor is influenced by its affinity for the receptor, which is a measure of how strongly and specifically the ligand binds to the receptor.

In competitive binding, if one ligand is present in high concentrations, it can prevent other ligands with lower affinity from binding to the receptor. This is because the higher-affinity ligand will have a greater probability of occupying the binding site and blocking access to the other ligands. The competition between ligands can be described mathematically using equations such as the Langmuir isotherm, which describes the relationship between the concentration of ligand and the fraction of receptors that are occupied by the ligand.

Competitive binding is an important concept in drug development, as it can be used to predict how different drugs will interact with their targets and how they may affect each other's activity. By understanding the competitive binding properties of a drug, researchers can optimize its dosage and delivery to maximize its therapeutic effect while minimizing unwanted side effects.

The pituitary gland is a small, endocrine gland located at the base of the brain, in the sella turcica of the sphenoid bone. It is often called the "master gland" because it controls other glands and makes the hormones that trigger many body functions. The pituitary gland measures about 0.5 cm in height and 1 cm in width, and it weighs approximately 0.5 grams.

The pituitary gland is divided into two main parts: the anterior lobe (adenohypophysis) and the posterior lobe (neurohypophysis). The anterior lobe is further divided into three zones: the pars distalis, pars intermedia, and pars tuberalis. Each part of the pituitary gland has distinct functions and produces different hormones.

The anterior pituitary gland produces and releases several important hormones, including:

* Growth hormone (GH), which regulates growth and development in children and helps maintain muscle mass and bone strength in adults.
* Thyroid-stimulating hormone (TSH), which controls the production of thyroid hormones by the thyroid gland.
* Adrenocorticotropic hormone (ACTH), which stimulates the adrenal glands to produce cortisol and other steroid hormones.
* Follicle-stimulating hormone (FSH) and luteinizing hormone (LH), which regulate reproductive function in both males and females.
* Prolactin, which stimulates milk production in pregnant and lactating women.

The posterior pituitary gland stores and releases two hormones that are produced by the hypothalamus:

* Antidiuretic hormone (ADH), which helps regulate water balance in the body by controlling urine production.
* Oxytocin, which stimulates uterine contractions during childbirth and milk release during breastfeeding.

Overall, the pituitary gland plays a critical role in maintaining homeostasis and regulating various bodily functions, including growth, development, metabolism, and reproductive function.

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.

HEK293 cells, also known as human embryonic kidney 293 cells, are a line of cells used in scientific research. They were originally derived from human embryonic kidney cells and have been adapted to grow in a lab setting. HEK293 cells are widely used in molecular biology and biochemistry because they can be easily transfected (a process by which DNA is introduced into cells) and highly express foreign genes. As a result, they are often used to produce proteins for structural and functional studies. It's important to note that while HEK293 cells are derived from human tissue, they have been grown in the lab for many generations and do not retain the characteristics of the original embryonic kidney cells.

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.

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.

Southern blotting is a type of membrane-based blotting technique that is used in molecular biology to detect and locate specific DNA sequences within a DNA sample. This technique is named after its inventor, Edward M. Southern.

In Southern blotting, the DNA sample is first digested with one or more restriction enzymes, which cut the DNA at specific recognition sites. The resulting DNA fragments are then separated based on their size by gel electrophoresis. After separation, the DNA fragments are denatured to convert them into single-stranded DNA and transferred onto a nitrocellulose or nylon membrane.

Once the DNA has been transferred to the membrane, it is hybridized with a labeled probe that is complementary to the sequence of interest. The probe can be labeled with radioactive isotopes, fluorescent dyes, or chemiluminescent compounds. After hybridization, the membrane is washed to remove any unbound probe and then exposed to X-ray film (in the case of radioactive probes) or scanned (in the case of non-radioactive probes) to detect the location of the labeled probe on the membrane.

The position of the labeled probe on the membrane corresponds to the location of the specific DNA sequence within the original DNA sample. Southern blotting is a powerful tool for identifying and characterizing specific DNA sequences, such as those associated with genetic diseases or gene regulation.

'Candida albicans' is a species of yeast that is commonly found in the human body, particularly in warm and moist areas such as the mouth, gut, and genital region. It is a part of the normal microbiota and usually does not cause any harm. However, under certain conditions like a weakened immune system, prolonged use of antibiotics or steroids, poor oral hygiene, or diabetes, it can overgrow and cause infections known as candidiasis. These infections can affect various parts of the body including the skin, nails, mouth (thrush), and genital area (yeast infection).

The medical definition of 'Candida albicans' is:

A species of yeast belonging to the genus Candida, which is commonly found as a commensal organism in humans. It can cause opportunistic infections when there is a disruption in the normal microbiota or when the immune system is compromised. The overgrowth of C. albicans can lead to various forms of candidiasis, such as oral thrush, vaginal yeast infection, and invasive candidiasis.

RNA splicing is a post-transcriptional modification process in which the non-coding sequences (introns) are removed and the coding sequences (exons) are joined together in a messenger RNA (mRNA) molecule. This results in a continuous mRNA sequence that can be translated into a single protein. Alternative splicing, where different combinations of exons are included or excluded, allows for the creation of multiple proteins from a single gene.

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.

Gene expression regulation in bacteria refers to the complex cellular processes that control the production of proteins from specific genes. This regulation allows bacteria to adapt to changing environmental conditions and ensure the appropriate amount of protein is produced at the right time.

Bacteria have a variety of mechanisms for regulating gene expression, including:

1. Operon structure: Many bacterial genes are organized into operons, which are clusters of genes that are transcribed together as a single mRNA molecule. The expression of these genes can be coordinately regulated by controlling the transcription of the entire operon.
2. Promoter regulation: Transcription is initiated at promoter regions upstream of the gene or operon. Bacteria have regulatory proteins called sigma factors that bind to the promoter and recruit RNA polymerase, the enzyme responsible for transcribing DNA into RNA. The binding of sigma factors can be influenced by environmental signals, allowing for regulation of transcription.
3. Attenuation: Some operons have regulatory regions called attenuators that control transcription termination. These regions contain hairpin structures that can form in the mRNA and cause transcription to stop prematurely. The formation of these hairpins is influenced by the concentration of specific metabolites, allowing for regulation of gene expression based on the availability of those metabolites.
4. Riboswitches: Some bacterial mRNAs contain regulatory elements called riboswitches that bind small molecules directly. When a small molecule binds to the riboswitch, it changes conformation and affects transcription or translation of the associated gene.
5. CRISPR-Cas systems: Bacteria use CRISPR-Cas systems for adaptive immunity against viruses and plasmids. These systems incorporate short sequences from foreign DNA into their own genome, which can then be used to recognize and cleave similar sequences in invading genetic elements.

Overall, gene expression regulation in bacteria is a complex process that allows them to respond quickly and efficiently to changing environmental conditions. Understanding these regulatory mechanisms can provide insights into bacterial physiology and help inform strategies for controlling bacterial growth and behavior.

DNA repair is the process by which cells identify and correct damage to the DNA molecules that encode their genome. DNA can be damaged by a variety of internal and external factors, such as radiation, chemicals, and metabolic byproducts. If left unrepaired, this damage can lead to mutations, which may in turn lead to cancer and other diseases.

There are several different mechanisms for repairing DNA damage, including:

1. Base excision repair (BER): This process repairs damage to a single base in the DNA molecule. An enzyme called a glycosylase removes the damaged base, leaving a gap that is then filled in by other enzymes.
2. Nucleotide excision repair (NER): This process repairs more severe damage, such as bulky adducts or crosslinks between the two strands of the DNA molecule. An enzyme cuts out a section of the damaged DNA, and the gap is then filled in by other enzymes.
3. Mismatch repair (MMR): This process repairs errors that occur during DNA replication, such as mismatched bases or small insertions or deletions. Specialized enzymes recognize the error and remove a section of the newly synthesized strand, which is then replaced by new nucleotides.
4. Double-strand break repair (DSBR): This process repairs breaks in both strands of the DNA molecule. There are two main pathways for DSBR: non-homologous end joining (NHEJ) and homologous recombination (HR). NHEJ directly rejoins the broken ends, while HR uses a template from a sister chromatid to repair the break.

Overall, DNA repair is a crucial process that helps maintain genome stability and prevent the development of diseases caused by genetic mutations.

Serine proteinase inhibitors, also known as serine protease inhibitors or serpins, are a group of proteins that inhibit serine proteases, which are enzymes that cut other proteins in a process called proteolysis. Serine proteinases are important in many biological processes such as blood coagulation, fibrinolysis, inflammation and cell death. The inhibition of these enzymes by serpin proteins is an essential regulatory mechanism to maintain the balance and prevent uncontrolled proteolytic activity that can lead to diseases.

Serpins work by forming a covalent complex with their target serine proteinases, irreversibly inactivating them. The active site of serpins contains a reactive center loop (RCL) that mimics the protease's target protein sequence and acts as a bait for the enzyme. When the protease cleaves the RCL, it gets trapped within the serpin structure, leading to its inactivation.

Serpin proteinase inhibitors play crucial roles in various physiological processes, including:

1. Blood coagulation and fibrinolysis regulation: Serpins such as antithrombin, heparin cofactor II, and protease nexin-2 control the activity of enzymes involved in blood clotting and dissolution to prevent excessive or insufficient clot formation.
2. Inflammation modulation: Serpins like α1-antitrypsin, α2-macroglobulin, and C1 inhibitor regulate the activity of proteases released during inflammation, protecting tissues from damage.
3. Cell death regulation: Some serpins, such as PI-9/SERPINB9, control apoptosis (programmed cell death) by inhibiting granzyme B, a protease involved in this process.
4. Embryonic development and tissue remodeling: Serpins like plasminogen activator inhibitor-1 (PAI-1) and PAI-2 regulate the activity of enzymes involved in extracellular matrix degradation during embryonic development and tissue remodeling.
5. Neuroprotection: Serpins such as neuroserpin protect neurons from damage by inhibiting proteases released during neuroinflammation or neurodegenerative diseases.

Dysregulation of serpins has been implicated in various pathological conditions, including thrombosis, emphysema, Alzheimer's disease, and cancer. Understanding the roles of serpins in these processes may provide insights into potential therapeutic strategies for treating these diseases.

HeLa cells are a type of immortalized cell line used in scientific research. They are derived from a cancer that developed in the cervical tissue of Henrietta Lacks, an African-American woman, in 1951. After her death, cells taken from her tumor were found to be capable of continuous division and growth in a laboratory setting, making them an invaluable resource for medical research.

HeLa cells have been used in a wide range of scientific studies, including research on cancer, viruses, genetics, and drug development. They were the first human cell line to be successfully cloned and are able to grow rapidly in culture, doubling their population every 20-24 hours. This has made them an essential tool for many areas of biomedical research.

It is important to note that while HeLa cells have been instrumental in numerous scientific breakthroughs, the story of their origin raises ethical questions about informed consent and the use of human tissue in research.

Heparin antagonists, also known as heparin neutralizers or reversal agents, are medications used to reverse the anticoagulant effects of heparin, a type of blood thinner. Heparin works by activating antithrombin III, which inactivates clotting factors IIa and Xa. Heparin antagonists, such as protamine sulfate, work by binding to heparin, forming a stable complex that is unable to bind to and activate antithrombin III, thereby neutralizing its anticoagulant effect.

Protamine sulfate is the most commonly used heparin antagonist. It is a highly basic protein derived from fish sperm that can neutralize the anticoagulant effects of heparin by forming a stable complex with it. The dose of protamine required to reverse the effects of heparin depends on the amount and type of heparin administered, as well as the timing of administration.

It is important to note that while heparin antagonists can reverse the anticoagulant effects of heparin, they do not reverse the underlying coagulation disorder or prevent further clot formation. Therefore, additional treatments may be necessary to manage the underlying condition and prevent recurrent thrombosis.

A "gene library" is not a recognized term in medical genetics or molecular biology. However, the closest concept that might be referred to by this term is a "genomic library," which is a collection of DNA clones that represent the entire genetic material of an organism. These libraries are used for various research purposes, such as identifying and studying specific genes or gene functions.

A missense mutation is a type of point mutation in which a single nucleotide change results in the substitution of a different amino acid in the protein that is encoded by the affected gene. This occurs when the altered codon (a sequence of three nucleotides that corresponds to a specific amino acid) specifies a different amino acid than the original one. The function and/or stability of the resulting protein may be affected, depending on the type and location of the missense mutation. Missense mutations can have various effects, ranging from benign to severe, depending on the importance of the changed amino acid for the protein's structure or function.

Experimental arthritis refers to the induction of joint inflammation in animal models for the purpose of studying the disease process and testing potential treatments. This is typically achieved through the use of various methods such as injecting certain chemicals or proteins into the joints, genetically modifying animals to develop arthritis-like symptoms, or immunizing animals to induce an autoimmune response against their own joint tissues. These models are crucial for advancing our understanding of the underlying mechanisms of arthritis and for developing new therapies to treat this debilitating disease.

Neurosecretory systems are specialized components of the nervous system that produce and release chemical messengers called neurohormones. These neurohormones are released into the bloodstream and can have endocrine effects on various target organs in the body. The cells that make up neurosecretory systems, known as neurosecretory cells, are found in specific regions of the brain, such as the hypothalamus, and in peripheral nerves.

Neurosecretory systems play a critical role in regulating many physiological processes, including fluid and electrolyte balance, stress responses, growth and development, reproductive functions, and behavior. The neurohormones released by these systems can act synergistically or antagonistically to maintain homeostasis and coordinate the body's response to internal and external stimuli.

Neurosecretory cells are characterized by their ability to synthesize and store neurohormones in secretory granules, which are released upon stimulation. The release of neurohormones can be triggered by a variety of signals, including neural impulses, hormonal changes, and other physiological cues. Once released into the bloodstream, neurohormones can travel to distant target organs, where they bind to specific receptors and elicit a range of responses.

Overall, neurosecretory systems are an essential component of the neuroendocrine system, which plays a critical role in regulating many aspects of human physiology and behavior.

Transaminases, also known as aminotransferases, are a group of enzymes found in various tissues of the body, particularly in the liver, heart, muscle, and kidneys. They play a crucial role in the metabolism of amino acids, the building blocks of proteins.

There are two major types of transaminases: aspartate aminotransferase (AST) and alanine aminotransferase (ALT). Both enzymes are normally present in low concentrations in the bloodstream. However, when tissues that contain these enzymes are damaged or injured, such as during liver disease or muscle damage, the levels of AST and ALT in the blood may significantly increase.

Measurement of serum transaminase levels is a common laboratory test used to assess liver function and detect liver injury or damage. Increased levels of these enzymes in the blood can indicate conditions such as hepatitis, liver cirrhosis, drug-induced liver injury, heart attack, and muscle disorders. It's important to note that while elevated transaminase levels may suggest liver disease, they do not specify the type or cause of the condition, and further diagnostic tests are often required for accurate diagnosis and treatment.

Amino acid motifs are recurring patterns or sequences of amino acids in a protein molecule. These motifs can be identified through various sequence analysis techniques and often have functional or structural significance. They can be as short as two amino acids in length, but typically contain at least three to five residues.

Some common examples of amino acid motifs include:

1. Active site motifs: These are specific sequences of amino acids that form the active site of an enzyme and participate in catalyzing chemical reactions. For example, the catalytic triad in serine proteases consists of three residues (serine, histidine, and aspartate) that work together to hydrolyze peptide bonds.
2. Signal peptide motifs: These are sequences of amino acids that target proteins for secretion or localization to specific organelles within the cell. For example, a typical signal peptide consists of a positively charged n-region, a hydrophobic h-region, and a polar c-region that directs the protein to the endoplasmic reticulum membrane for translocation.
3. Zinc finger motifs: These are structural domains that contain conserved sequences of amino acids that bind zinc ions and play important roles in DNA recognition and regulation of gene expression.
4. Transmembrane motifs: These are sequences of hydrophobic amino acids that span the lipid bilayer of cell membranes and anchor transmembrane proteins in place.
5. Phosphorylation sites: These are specific serine, threonine, or tyrosine residues that can be phosphorylated by protein kinases to regulate protein function.

Understanding amino acid motifs is important for predicting protein structure and function, as well as for identifying potential drug targets in disease-associated proteins.

Hyperlipoproteinemia Type II, also known as Fredrickson Type II or Familial Combined Hyperlipidemia, is a genetic disorder characterized by elevated levels of low-density lipoprotein (LDL) cholesterol and/or triglycerides in the blood. This condition can lead to an increased risk of developing cardiovascular diseases such as atherosclerosis and coronary artery disease.

The disorder is caused by mutations in several genes involved in lipid metabolism, including APOB, LDLR, PCSK9, and APOE. These genetic defects result in impaired clearance of LDL particles from the bloodstream, leading to their accumulation and increased risk of cardiovascular disease.

Individuals with Hyperlipoproteinemia Type II typically have elevated levels of both LDL cholesterol and triglycerides, although some may only have one or the other elevated. The disorder can present at any age, but it is often diagnosed in adulthood during routine cholesterol screening.

Treatment for Hyperlipoproteinemia Type II typically involves lifestyle modifications such as a heart-healthy diet, regular exercise, and weight loss. Medications such as statins, ezetimibe, and PCSK9 inhibitors may also be prescribed to lower LDL cholesterol levels and reduce the risk of cardiovascular disease.

Cathepsin H is a lysosomal cysteine protease that plays a role in intracellular protein degradation and turnover. It is expressed in various tissues, including the spleen, thymus, lungs, and immune cells. Cathepsin H has been implicated in several physiological processes, such as antigen presentation, bone resorption, and extracellular matrix remodeling. Additionally, its dysregulation has been associated with various pathological conditions, including cancer, neurodegenerative disorders, and infectious diseases.

The enzyme's active site contains a catalytic triad composed of cysteine, histidine, and aspartic acid residues, which facilitates the proteolytic activity. Cathepsin H exhibits specificity for peptide bonds containing hydrophobic or aromatic amino acids, making it an important player in processing and degrading various cellular proteins.

In summary, Cathepsin H is a lysosomal cysteine protease involved in protein turnover and degradation with potential implications in several pathological conditions when dysregulated.

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.

A point mutation is a type of genetic mutation where a single nucleotide base (A, T, C, or G) in DNA is altered, deleted, or substituted with another nucleotide. Point mutations can have various effects on the organism, depending on the location of the mutation and whether it affects the function of any genes. Some point mutations may not have any noticeable effect, while others might lead to changes in the amino acids that make up proteins, potentially causing diseases or altering traits. Point mutations can occur spontaneously due to errors during DNA replication or be inherited from parents.

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.

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.

A sequence deletion in a genetic context refers to the removal or absence of one or more nucleotides (the building blocks of DNA or RNA) from a specific region in a DNA or RNA molecule. This type of mutation can lead to the loss of genetic information, potentially resulting in changes in the function or expression of a gene. If the deletion involves a critical portion of the gene, it can cause diseases, depending on the role of that gene in the body. The size of the deleted sequence can vary, ranging from a single nucleotide to a large segment of DNA.

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.

Snake venoms are complex mixtures of bioactive compounds produced by specialized glands in snakes. They primarily consist of proteins and peptides, including enzymes, neurotoxins, hemotoxins, cytotoxins, and cardiotoxins. These toxins can cause a variety of pharmacological effects on the victim's body, such as disruption of the nervous system, blood coagulation, muscle function, and cell membrane integrity, ultimately leading to tissue damage and potentially death. The composition of snake venoms varies widely among different species, making each species' venom unique in its toxicity profile.

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

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

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

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

Proteinuria is a medical term that refers to the presence of excess proteins, particularly albumin, in the urine. Under normal circumstances, only small amounts of proteins should be found in the urine because the majority of proteins are too large to pass through the glomeruli, which are the filtering units of the kidneys.

However, when the glomeruli become damaged or diseased, they may allow larger molecules such as proteins to leak into the urine. Persistent proteinuria is often a sign of kidney disease and can indicate damage to the glomeruli. It is usually detected through a routine urinalysis and may be confirmed with further testing.

The severity of proteinuria can vary, and it can be a symptom of various underlying conditions such as diabetes, hypertension, glomerulonephritis, and other kidney diseases. Treatment for proteinuria depends on the underlying cause and may include medications to control blood pressure, manage diabetes, or reduce protein loss in the urine.

RNA splice sites are specific sequences on the pre-messenger RNA (pre-mRNA) molecule where the splicing process occurs during gene expression in eukaryotic cells. The pre-mRNA contains introns and exons, which are non-coding and coding regions of the RNA, respectively.

The splicing process removes the introns and joins together the exons to form a mature mRNA molecule that can be translated into a protein. The splice sites are recognized by the spliceosome, a complex of proteins and small nuclear RNAs (snRNAs) that catalyze the splicing reaction.

There are two main types of splice sites: the 5' splice site and the 3' splice site. The 5' splice site is located at the junction between the 5' end of the intron and the 3' end of the exon, while the 3' splice site is located at the junction between the 3' end of the intron and the 5' end of the exon.

The 5' splice site contains a conserved GU sequence, while the 3' splice site contains a conserved AG sequence. These sequences are recognized by the snRNAs in the spliceosome, which bind to them and facilitate the splicing reaction.

Mutations or variations in RNA splice sites can lead to abnormal splicing and result in diseases such as cancer, neurodegenerative disorders, and genetic disorders.

Chemical precipitation is a process in which a chemical compound becomes a solid, insoluble form, known as a precipitate, from a liquid solution. This occurs when the concentration of the compound in the solution exceeds its solubility limit and forms a separate phase. The reaction that causes the formation of the precipitate can be a result of various factors such as changes in temperature, pH, or the addition of another chemical reagent.

In the medical field, chemical precipitation is used in diagnostic tests to detect and measure the presence of certain substances in body fluids, such as blood or urine. For example, a common test for kidney function involves adding a chemical reagent to a urine sample, which causes the excess protein in the urine to precipitate out of solution. The amount of precipitate formed can then be measured and used to diagnose and monitor kidney disease.

Chemical precipitation is also used in the treatment of certain medical conditions, such as heavy metal poisoning. In this case, a chelating agent is administered to bind with the toxic metal ions in the body, forming an insoluble compound that can be excreted through the urine or feces. This process helps to reduce the amount of toxic metals in the body and alleviate symptoms associated with poisoning.

Enkephalins are naturally occurring opioid peptides that bind to opiate receptors in the brain and other organs, producing pain-relieving and other effects. They are derived from the precursor protein proenkephalin and consist of two main types: Leu-enkephalin and Met-enkephalin. Enkephalins play a role in pain modulation, stress response, mood regulation, and addictive behaviors. They are also involved in the body's reward system and have been implicated in various physiological processes such as respiration, gastrointestinal motility, and hormone release.

Reperfusion injury is a complex pathophysiological process that occurs when blood flow is restored to previously ischemic tissues, leading to further tissue damage. This phenomenon can occur in various clinical settings such as myocardial infarction (heart attack), stroke, or peripheral artery disease after an intervention aimed at restoring perfusion.

The restoration of blood flow leads to the generation of reactive oxygen species (ROS) and inflammatory mediators, which can cause oxidative stress, cellular damage, and activation of the immune system. This results in a cascade of events that may lead to microvascular dysfunction, capillary leakage, and tissue edema, further exacerbating the injury.

Reperfusion injury is an important consideration in the management of ischemic events, as interventions aimed at restoring blood flow must be carefully balanced with potential harm from reperfusion injury. Strategies to mitigate reperfusion injury include ischemic preconditioning (exposing the tissue to short periods of ischemia before a prolonged ischemic event), ischemic postconditioning (applying brief periods of ischemia and reperfusion after restoring blood flow), remote ischemic preconditioning (ischemia applied to a distant organ or tissue to protect the target organ), and pharmacological interventions that scavenge ROS, reduce inflammation, or improve microvascular function.

The cell nucleus is a membrane-bound organelle found in the eukaryotic cells (cells with a true nucleus). It contains most of the cell's genetic material, organized as DNA molecules in complex with proteins, RNA molecules, and histones to form chromosomes.

The primary function of the cell nucleus is to regulate and control the activities of the cell, including growth, metabolism, protein synthesis, and reproduction. It also plays a crucial role in the process of mitosis (cell division) by separating and protecting the genetic material during this process. The nuclear membrane, or nuclear envelope, surrounding the nucleus is composed of two lipid bilayers with numerous pores that allow for the selective transport of molecules between the nucleoplasm (nucleus interior) and the cytoplasm (cell exterior).

The cell nucleus is a vital structure in eukaryotic cells, and its dysfunction can lead to various diseases, including cancer and genetic disorders.

In the context of medical research, "methods" refers to the specific procedures or techniques used in conducting a study or experiment. This includes details on how data was collected, what measurements were taken, and what statistical analyses were performed. The methods section of a medical paper allows other researchers to replicate the study if they choose to do so. It is considered one of the key components of a well-written research article, as it provides transparency and helps establish the validity of the findings.

In situ hybridization (ISH) is a molecular biology technique used to detect and localize specific nucleic acid sequences, such as DNA or RNA, within cells or tissues. This technique involves the use of a labeled probe that is complementary to the target nucleic acid sequence. The probe can be labeled with various types of markers, including radioisotopes, fluorescent dyes, or enzymes.

During the ISH procedure, the labeled probe is hybridized to the target nucleic acid sequence in situ, meaning that the hybridization occurs within the intact cells or tissues. After washing away unbound probe, the location of the labeled probe can be visualized using various methods depending on the type of label used.

In situ hybridization has a wide range of applications in both research and diagnostic settings, including the detection of gene expression patterns, identification of viral infections, and diagnosis of genetic disorders.

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.

Transcription factor RelB is a member of the NF-κB (nuclear factor kappa B) family, which plays a crucial role in regulating immune responses, cell survival, and inflammation. RelB forms a heterodimer with other NF-κB family members, such as p50 or p52, and binds to specific DNA sequences called κB sites in the promoter regions of target genes. This binding leads to the activation or repression of gene transcription, ultimately influencing various cellular processes, including immune response regulation, development, and oncogenesis. RelB is unique among NF-κB family members because it can shuttle between the cytoplasm and nucleus even in unstimulated cells, although its activity is enhanced upon stimulation by various signals.

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

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

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

'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.

Fungal proteins are a type of protein that is specifically produced and present in fungi, which are a group of eukaryotic organisms that include microorganisms such as yeasts and molds. These proteins play various roles in the growth, development, and survival of fungi. They can be involved in the structure and function of fungal cells, metabolism, pathogenesis, and other cellular processes. Some fungal proteins can also have important implications for human health, both in terms of their potential use as therapeutic targets and as allergens or toxins that can cause disease.

Fungal proteins can be classified into different categories based on their functions, such as enzymes, structural proteins, signaling proteins, and toxins. Enzymes are proteins that catalyze chemical reactions in fungal cells, while structural proteins provide support and protection for the cell. Signaling proteins are involved in communication between cells and regulation of various cellular processes, and toxins are proteins that can cause harm to other organisms, including humans.

Understanding the structure and function of fungal proteins is important for developing new treatments for fungal infections, as well as for understanding the basic biology of fungi. Research on fungal proteins has led to the development of several antifungal drugs that target specific fungal enzymes or other proteins, providing effective treatment options for a range of fungal diseases. Additionally, further study of fungal proteins may reveal new targets for drug development and help improve our ability to diagnose and treat fungal infections.

Neuraminidase is an enzyme that occurs on the surface of influenza viruses. It plays a crucial role in the life cycle of the virus by helping it to infect host cells and to spread from cell to cell within the body. Neuraminidase works by cleaving sialic acid residues from glycoproteins, allowing the virus to detach from infected cells and to move through mucus and other bodily fluids. This enzyme is a major target of antiviral drugs used to treat influenza, such as oseltamivir (Tamiflu) and zanamivir (Relenza). Inhibiting the activity of neuraminidase can help to prevent the spread of the virus within the body and reduce the severity of symptoms.

Amino acids are organic compounds that serve as the building blocks of proteins. They consist of a central carbon atom, also known as the alpha carbon, which is bonded to an amino group (-NH2), a carboxyl group (-COOH), a hydrogen atom (H), and a variable side chain (R group). The R group can be composed of various combinations of atoms such as hydrogen, oxygen, sulfur, nitrogen, and carbon, which determine the unique properties of each amino acid.

There are 20 standard amino acids that are encoded by the genetic code and incorporated into proteins during translation. These include:

1. Alanine (Ala)
2. Arginine (Arg)
3. Asparagine (Asn)
4. Aspartic acid (Asp)
5. Cysteine (Cys)
6. Glutamine (Gln)
7. Glutamic acid (Glu)
8. Glycine (Gly)
9. Histidine (His)
10. Isoleucine (Ile)
11. Leucine (Leu)
12. Lysine (Lys)
13. Methionine (Met)
14. Phenylalanine (Phe)
15. Proline (Pro)
16. Serine (Ser)
17. Threonine (Thr)
18. Tryptophan (Trp)
19. Tyrosine (Tyr)
20. Valine (Val)

Additionally, there are several non-standard or modified amino acids that can be incorporated into proteins through post-translational modifications, such as hydroxylation, methylation, and phosphorylation. These modifications expand the functional diversity of proteins and play crucial roles in various cellular processes.

Amino acids are essential for numerous biological functions, including protein synthesis, enzyme catalysis, neurotransmitter production, energy metabolism, and immune response regulation. Some amino acids can be synthesized by the human body (non-essential), while others must be obtained through dietary sources (essential).

I must clarify that the term "pedigree" is not typically used in medical definitions. Instead, it is often employed in genetics and breeding, where it refers to the recorded ancestry of an individual or a family, tracing the inheritance of specific traits or diseases. In human genetics, a pedigree can help illustrate the pattern of genetic inheritance in families over multiple generations. However, it is not a medical term with a specific clinical definition.

... two regulators of complement. The membrane attack complex is initiated when the complement protein C5 convertase cleaves C5 ... All three pathways of the complement system (classical, lectin and alternative pathways) initiate the formation of MAC. Another ... Terminal complement pathway deficiency Paroxysmal nocturnal haemoglobinuria Perforin Pore-forming toxin Xie CB, Jane-Wit D, ... ISBN 978-0-323-54943-1. Media related to Complement membrane attack complex at Wikimedia Commons Complement+Membrane+Attack+ ...
... a novel C3d-targeted C3/C5 convertase inhibitor for treatment of human complement alternative pathway-mediated diseases". Blood ... A suggested alternative to linkage studies was the genetic association study. This study type asks if the allele of a genetic ... A common alternative to case-control GWA studies is the analysis of quantitative phenotypic data, e.g. height or biomarker ... An alternative application is therefore the potential for GWA studies to elucidate pathophysiology. One such success is related ...
... a stable compound which can bind an additional C3b to form alternative pathway C5-convertase. The C5-convertase of the ... The alternative pathway is one of three complement pathways that opsonize and kill pathogens. The pathway is triggered when the ... This complex is also known as a fluid-phase C3-convertase. This convertase, the alternative pathway C3-convertase, although ... After the creation of C5 convertase (either as (C3b)2BbP or C4b2b3b from the classical pathway), the complement system follows ...
The term C3 convertase may refer to: C3-convertase, an enzyme Alternative-complement-pathway C3/C5 convertase, an enzyme This ...
C3b binds to the C3 convertase (C4b2b), to form C5 convertase (C4b2b3b). C5 convertase then cleaves C5 into C5a and C5b. Like ... Alternative complement pathway - another complement system pathway Lectin pathway - another complement system pathway Noris, ... Activation of the complement pathway through the classical, lectin or alternative complement pathway is followed by a cascade ... The classical complement pathway is one of three pathways which activate the complement system, which is part of the immune ...
... or the alternative pathway (C3bBbC3b) of complement system. Two fluid phase C5 convertases have been described: the classical ... The target of C5 convertase is complement protein C5. C5 is a two-chain (α, β) plasma glycoprotein (Mr = 196,000). C5 and C3 ... The complement component C5 can be also activated by fluid phase C5 convertase. C5 is activated by CVFBb in the presence of ... "Alternative pathway of complement: Recruitment of precursor properdin by the labile C3/C5 convertase and the potentiation of ...
... complement factor I EC 3.4.21.46: complement factor D EC 3.4.21.47: alternative-complement-pathway C3/C5 convertase EC 3.4. ... classical-complement-pathway C3/C5 convertase EC 3.4.21.44: Now EC 3.4.21.43, classical-complement-pathway C3/C5 convertase EC ... proprotein convertase 1 EC 3.4.21.94: proprotein convertase 2 EC 3.4.21.95: snake venom factor V activator EC 3.4.21.96: ... Complement subcomponent C1r EC 3.4.21.42: complement subcomponent C1s EC 3.4.21.43: ...
C5 convertase formation and cell lysis. C3 convertase can be used to refer to the form produced in the alternative pathway ( ... Hourcade D (2006). "The Role of Properdin in the Assembly of the Alternative Pathway C3 Convertases of Complement". J Biol Chem ... It is a membrane protein and regulates also C5 convertase of the classical and alternative pathway. DAF protects host cells ... Since C3 convertases cleave C3 to produce C3b which can then form an additional C3 convertase through the alternative pathway, ...
C3 nephritic factor is an autoantibody to the complement C3bBb convertase that activates the alternative complement pathway, a ... finding that suggested the potential of B cell or complement C5 depletion as adjunct therapies in certain forms of kidney ...
... iC3b C5 - C5a C3-convertase C5-convertase Late stage Membrane attack complex (MAC) C6 C7 C8 C9 Complement pathway inhibitors C1 ... C4a C2 Mannan-binding lectin pathway MASP1 / MASP2 Mannan-binding lectin Alternative complement pathway Factor B Factor D ... system Complement system Classical complement pathway Mannan-binding lectin pathway Alternate complement pathway Complement ... divided by pathway) Classical complement pathway C1Q complex - C1R / C1S C4 - ...
... components of the alternative-pathway C3 convertase of complement". The Biochemical Journal. 253 (3): 667-75. doi:10.1042/ ... Rawal N, Pangburn MK (March 2001). "Structure/function of C5 convertases of complement". International Immunopharmacology. 1 (3 ... This gene encodes complement factor B, a component of the alternative pathway of complement activation. Factor B circulates in ... The active subunit Bb is a serine protease that associates with C3b to form the alternative pathway C3 convertase. Bb is ...
Hourcade D (2006). "The Role of Properdin in the Assembly of the Alternative Pathway C3 Convertases of Complement". J Biol Chem ... This protein binds to bacterial cell walls and dying human cells to stabilize the C3 and C5-convertase enzyme complexes to form ... It binds to preformed alternative pathway C3-convertases. Properdin also inhibits the Factor H - mediated cleavage of C3b by ... The alternative pathway is not dependent on antibodies. This branch of the complement system is activated by IgA immune ...
... alternative pathway and lectin pathway) that ultimately lead to the formation of a C3 convertase. Formation of a C3 convertase ... In such cases treatment with the complement-inhibitory anti-C5 monoclonal antibody, eculizumab, is found to be highly effective ... Additionally, C3b plays a role in forming a C3 convertase when bound to Factor B (C3bBb complex), or a C5 convertase when bound ... Binding of a C3b molecule to the C4bC2b complex (C4b2b3b) results in the formation of a C5 convertase, which cleaves C5 into ...
C3b later joins with C4b2b to make C5 convertase (C4b2b3b complex). The alternative pathway is continuously activated at a low ... the classical complement pathway, the alternative complement pathway, and the lectin pathway. The alternative pathway accounts ... the alternative complement pathway is one element of innate immunity.[citation needed] Once the alternative C3 convertase ... The complex of C3b(2)Bb is a protease which cleaves C5 into C5b and C5a. C5 convertase is also formed by the classical pathway ...
... participates in all three of the complement pathways (classical, alternative, and lectin); the alternative pathway is " ... C5 binding sites, 4) private allelic residues. Additionally, the same study identified the expression of human complement C4 ... the C4b-C2a complex with protease activity has been termed the C3 convertase. Protein 4b can be further cleaved into 4c and 4d ... Complement component 4A Complement component 4B HLA A1-B8-DR3-DQ2 haplotype Complement system Complement deficiency Sekar A, ...
... complement c3-c5 convertases MeSH D12.776.124.486.274.860.387.500 - complement c3-c5 convertases, alternative pathway MeSH ... complement c5 convertase, alternative pathway MeSH D12.776.124.486.274.860.387.750 - complement c3-c5 convertases, classical ... complement c3 convertase, classical pathway MeSH D12.776.124.486.274.860.387.750.750 - complement c5 convertase, classical ... complement c3 convertase, alternative pathway MeSH D12.776.124.486.274.860.387.500.750 - ...
... amongst other complement proteins and factors, leading to regulation of the alternative pathway of complement in particular. ... and decay accelerating activity against the alternative pathway C3-convertase, C3bBb. Factor H exerts its protective action on ... January 2011). "The development of atypical hemolytic uremic syndrome depends on complement C5". Journal of the American ... Its principal function is to regulate the alternative pathway of the complement system, ensuring that the complement system is ...
... two regulators of complement. The membrane attack complex is initiated when the complement protein C5 convertase cleaves C5 ... All three pathways of the complement system (classical, lectin and alternative pathways) initiate the formation of MAC. Another ... Terminal complement pathway deficiency Paroxysmal nocturnal haemoglobinuria Perforin Pore-forming toxin Xie CB, Jane-Wit D, ... ISBN 978-0-323-54943-1. Media related to Complement membrane attack complex at Wikimedia Commons Complement+Membrane+Attack+ ...
C5-9).The IgG antibody, C3NeF, prevents the alternative complement C3-convertase C3Bb from dissociative inactivation, resulting ... Adipocytes synthesize factor D, the limiting component of the alternative complement pathway, which cleaves C3-bound factor B ... Activation of alternate complement pathway has been demonstrated in most patients. The phenotype can be variable and sometimes ... in which subendothelial deposits of immunoglobulin and C3 are probably due to a deregulated alternative complement pathway. ...
Alternative Complement Pathway C3 C5 Convertase 66% 9 Citations (Scopus) 51 Downloads (Pure) ... Adoption of in vitro systems and zebrafish embryos as alternative models for reducing rodent use in assessments of ... Transgenic zebrafish larvae as a non-rodent alternative model to assess pro-inflammatory (neutrophil) responses to ...
Alternative Complement Pathway C3 C5 Convertase 100% * Cardiac Graft Rejection 100% * Hemeprotein 100% ... Human T-cell lymphotropic virus type 1 Tax1 activation of NF-κB: Involvement of the protein kinase C pathway. Lindholm, P. F., ...
Activation of the terminal complement pathway occurs when the alternative and classical C5 convertases C3bBbC3b or C4b2aC3b ... FB is activated by factor D forming the alternative pathway C3 convertase. The alternative pathway also amplifies the classical ... C3 is cleaved to C3a and C3b to form the C5 convertase C4b2aC3b. The lectin complement pathway is homologous to the classical ... C3 is subsequently cleaved to C3a and C3b to form the C5 convertase C3BbC3b. Activation of the classical complement pathway ...
Alternative Complement Pathway C3 C5 Convertase Pharmacology, Toxicology and Pharmaceutical Science 85% ... cigarettes promoted cancer stem-like cell generation in the head and neck area via the OCT-4/ID1/NF-κB/BMI-1 signaling pathway. ... cigarettes promoted cancer stem-like cell generation in the head and neck area via the OCT-4/ID1/NF-κB/BMI-1 signaling pathway. ... cigarettes promoted cancer stem-like cell generation in the head and neck area via the OCT-4/ID1/NF-κB/BMI-1 signaling pathway. ...
Alternative Complement Pathway C3 C5 Convertase 11% * Fat 100% * Glucose Transport 33% ... Dive into the research topics of Effects of a brief high-fat diet and acute exercise on the mTORC1 and IKK/NF-kB pathways in ... Effects of a brief high-fat diet and acute exercise on the mTORC1 and IKK/NF-kB pathways in rat skeletal muscle. ...
Alternative Complement Pathway C3 C5 Convertase 11% * Interleukin 1 11% * Side Effect 11% ...
Alternative Complement Pathway C3 C5 Convertase 10% * Nuclear Factor 10% * Protein 10% ...
Alternative Complement Pathway C3 C5 Convertase 100% * Transcription Factor RUNX2 100% * Aorta 74% ...
Alternative Complement Pathway C3 C5 Convertase 100% * Nuclear Factor 100% 60 Scopus citations ... PTEN and TNF-α regulation of the intestinal-specific Cdx-2 homeobox gene through a PI3K, PKB/Akt, and NF-κB-dependent pathway. ... Regulation of TRAIL expression by the phosphatidylinositol 3-kinase/Akt/GSK-3 pathway in human colon cancer cells. Wang, Q., ... Selective targeting of the nuclear factor-κB pathway enhances tumor necrosis factor-related apoptosis-inducing ligand-mediated ...
Alternative Complement Pathway C3 C5 Convertase 50% * Nicotinamide Adenine Dinucleotide Adenosine Diphosphate ...
Alternative Complement Pathway C3 C5 Convertase Neuroscience 28% * Nuclear Factor Neuroscience 28% ... signaling pathways were determined by western immunoblotting. The function of mitogen-activated protein kinase phosphatase 1 ( ... signaling pathways were determined by western immunoblotting. The function of mitogen-activated protein kinase phosphatase 1 ( ... signaling pathways were determined by western immunoblotting. The function of mitogen-activated protein kinase phosphatase 1 ( ...
C5-9).The IgG antibody, C3Nef, prevents the alternative complement C3-convertase C3Bb from dissociative inactivation, resulting ... C3 nephritic factor is a serum immunoglobulin G that interacts with the C3bBb alternative pathway convertase to activate C3. ... C5-9).The IgG antibody, C3Nef, prevents the alternative complement C3-convertase C3Bb from dissociative inactivation, resulting ... Adipocytes synthesize factor D, the limiting component of the alternative complement pathway, which cleaves C3-bound factor B ...
NFt stabilizes the alternative pathway properdin-dependent C3/C5 convertase (C3Bb2,Bb,P) and leads to C3 activation and ... NFa stabilizes the alternative pathway convertase and results in complement activation and chronic C3 consumption. Deficiency ... The alternative pathway utilizes C3 and factors B and D to form the alternative pathway convertase C3b,Bb. ... NFt stabilizes the alternative pathway properdin-dependent C3/C5 convertase (C3Bb2,Bb,P) and also activates the terminal ...
... which leads to the activation of the terminal complement pathway. In the terminal pathway, 2 types of C5 convertase (C4bC2C3b ... alternative pathway; CFHR: complement factor H-related; CP: classical pathway; CR1: complement receptor 1; FB: factor B; FD: ... NETosing neutrophils activate complement both on their own NETs and bacteria via alternative and non-alternative pathways. ... The addition of C3b to C3 convertase in all 3 pathways generates C5 convertase, ...
C5 convertase) in the alternative complement pathway.. Alternative products: 2 isoforms; 1 and 2; are produced by alternative ... Three-dimensional structure of a complement control protein module in solution.. 1HFH. Solution structure of a pair of ... complement modules by nuclear magnetic resonance.. 1HFI. Solution structure of a pair of complement modules by nuclear magnetic ... C3 convertase) and the (C3b)nbb complex ( ...
The local amplification process leads to the creation of the alternative pathway C5 convertase, C3bBb3b, and initiates the ... terminal pathway of complement activation. The alternative pathway may account for ca 80% of the terminal pathway activity. ... Positive regulator plasma protein properdin is critical for the alternative pathway of complement. It is a single-chain ... Properdin can both initiate and positively regulate the alternative pathway activity together with C3 and factors B, D, I and H ...
C5-9).The IgG antibody, C3Nef, prevents the alternative complement C3-convertase C3Bb from dissociative inactivation, resulting ... Adipocytes synthesize factor D, the limiting component of the alternative complement pathway, which cleaves C3-bound factor B ... Activation of alternate complement pathway has been demonstrated in most patients. The phenotype can be variable and sometimes ... in which subendothelial deposits of immunoglobulin and C3 are probably due to a deregulated alternative complement pathway. ...
C5-9).The IgG antibody, C3Nef, prevents the alternative complement C3-convertase C3Bb from dissociative inactivation, resulting ... Adipocytes synthesize factor D, the limiting component of the alternative complement pathway, which cleaves C3-bound factor B ... in which subendothelial deposits of immunoglobulin and C3 are probably due to a deregulated alternative complement pathway. ... but which usually does not result in the activation of the terminal lytic part of the complement pathway ( ...
Learn about the three pathways lead to complement activation and some of their key inhibitors. ... The latter binds to the C3 convertase complex to form C4b2a3b, the classical pathway C5 convertase. This complex cleaves C5 ... The alternative pathway. The alternative pathway of complement activation is in a constant state of low-level activation (known ... The latter binds to the C3 convertase complex to form C4b2a3b, the classical pathway C5 convertase. This complex cleaves C5 ...
The C5 convertase of the classical pathway, as well as the C3 and C5 convertases of the alternative pathways, contain at least ... Ricklin D. Manipulating the mediator: modulation of the alternative complement pathway C3 convertase in health, disease and ... Classical and alternative complement pathways. Healthy volunteer sera were incubated at 37°C for 1 hour with Gp96 produced in ... Gp96 inhibits the alternative and the classical pathway of complement C3 activation in human serum. Initiation of the ...
Factor B is one of the proteins unique to the Alternative Complement Pathway. ... Bb can then become the enzymatically active constituent of the C3 and C5 alternative pathway convertases. ... Purified human complement Factor B. Factor B is one of the proteins unique to the Alternative Complement Pathway. The 93 kD ... When the alternative pathway is activated either directly or by the classical pathway via the amplification loop, C3b can bind ...
This complex is a C3/C5 convertase that cleaves both complement components C3 and C5. Unlike endogenous C3b/Bb, CVF/Bb is a ... and alternative pathways), and its various activation products are crucial for mediating, directly or indirectly, virtually all ... complement component C3 is the most important protein of the complement system as it is activated by all three pathways ( ... In venom, complement C3 homolog is a structural and functional analog of complement component C3b, the activated form of C3. ...
... recognizes human complement factor H which exists in 2 forms. The most common form, of 150 kDa, and the les… ... strong,Mouse anti Human Complement Factor H antibody, clone 028B-244.2.10X (10-10),/strong, ... C5 convertase). This prevents these components of the classical and of alternative complement pathways from forming a positive ... It is important in regulating the complement pathway, preventing unnecessary inflammation which can damage the host tissue.. ...
... this complex will then be cleaved by factor D into Ba and the alternative pathway C3-convertase, Bb. In other words, after ... Furthermore, products of C3 and C5 enhance neutrophil phagocytosis, that is, they are chemokines. ... Three biochemical pathways activate the complement system: the classical complement pathway, the alternate complement pathway, ... Alternative pathway. The alternative pathway is triggered by C3 hydrolysis directly on the surface of a pathogen. It does not ...
The membrane-bound complement inhibitor CD55 acts on the C3/C5 convertase level. Here, we fused MAP-2 to the four N-terminal ... and alternative pathways, respectively), MAP-2:CD551-4 demonstrated a potent inhibitory activity (IC50MAP-2:CD55 1-4 = 2.94, ... While MAP-2 and CD55 alone showed a minor inhibition of the three complement pathways when co-incubated with serum (IC50MAP-2+ ... Thus, the development of complement modulators has risen as a potential treatment for complement-driven inflammatory ...
The Alternative Pathway. In a similar manner to the classical pathway, the alternative pathway works by forming the MAC ( ... The pathway can now continue like the classical pathway with the activation of C3, resulting in the activation of C5. The ... The now activated C3b acts as an enzyme (C3b convertase) for further recruitment of C3 ... There are three types of complement cascade, the classical and alternative pathways and the Mannan-binding lectin pathway. Both ...
Patients with aHUS typically have genetic or acquired defects in the alternative complement pathway (AP). The complement system ... including the formation of C5 convertase (C3bBbC3b) [11]. C5 convertase then generates C5a and C5b, and C5b ultimately binds to ... aHUS is associated with deregulated complement systems, and is treated with eculizumab, a terminal C5 complement inhibitor [1,2 ... the complement system is up-regulated, and this may unmask defective complement regulation [8]. Originally designed to treat ...
  • The membrane attack complex (MAC) or terminal complement complex (TCC) is a complex of proteins typically formed on the surface of pathogen cell membranes as a result of the activation of the host's complement system, and as such is an effector of the immune system. (wikipedia.org)
  • MAC is composed of a complex of four complement proteins (C5b, C6, C7, and C8) that bind to the outer surface of the plasma membrane, and many copies of a fifth protein (C9) that hook up to one another, forming a ring in the membrane. (wikipedia.org)
  • The levels of proteins in mitogen-activated protein kinases (MAPKs), nuclear factor erythroid 2-related factor 2 (Nrf2), and nuclear factor-κB (NF-κB) signaling pathways were determined by western immunoblotting. (uky.edu)
  • 10 This distinction is not absolute because genetic defects in complement proteins have been identified in secondary TMA. (jrheum.org)
  • Purified human complement Factor B. Factor B is one of the proteins unique to the Alternative Complement Pathway. (quidel.com)
  • The complement system consists of more than 35 soluble and cell-bound proteins, 12 of which are directly involved in the complement pathways. (dadamo.com)
  • Our results demonstrate that MAP-2:CD551-4 is a potent complement inhibitor reinforcing the idea that engineered fusion proteins are a promising design strategy for identifying and developing drug candidates to treat complement-mediated diseases. (bvsalud.org)
  • The complement system or complement cascade as it is also known is a complex system of multiple proteins involved in inflammation and immunological response. (vetsci.co.uk)
  • 2 The resulting C3bBb can bind to other C3 proteins, leading to a positive feedback loop of complement activation. (drugbank.com)
  • In HAE types I and II, complement proteins C2 and C4 are low. (mastattack.org)
  • In vivo, regulation of complement at the C3 and C5 activation steps is provided by both plasma and membrane proteins. (justia.com)
  • The plasma protein inhibitors are factor H and C4-binding protein, and the regulatory membrane proteins located on cell surfaces are complement receptors 1 (CR1), decay-accelerating factor (DAF), and membrane cofactor protein (MCP). (justia.com)
  • These proteins inhibit the C3 and C5 convertases (multi-subunit proteases), by promoting dissociation of the multisubunit complexes and/or by inactivating the complexes through proteolysis (catalyzed by factor I). Several pharmacological agents that regulate or modulate complement activity have been identified by in vitro assay, but most have been shown in vivo to be of low activity or toxic. (justia.com)
  • The complement system consists of a number of small proteins found in the blood, normally circulating as inactive zymogens . (wikidoc.org)
  • Over 20 proteins and protein fragments make up the complement system, including serum proteins, serosal proteins, and cell membrane receptors. (wikidoc.org)
  • E.g. loss of CFH inhibition (mutations in CFH or auto-antibodies to CFH), CFH deregulation (from altered CFHR proteins), autoantibodies that stabilize C3 convertase (C3 nephritic factor), or impaired inactivation of C3b. (glomcon.org)
  • This results in overactivity of C3 convertase and then C5 convertase, with resulting deposition of complement proteins in the glomerulus. (glomcon.org)
  • Enzymes that activate one or more COMPLEMENT PROTEINS in the complement system leading to the formation of the COMPLEMENT MEMBRANE ATTACK COMPLEX, an important response in host defense. (lookformedical.com)
  • In blood, complement proteins are a major component and perhaps therefore most of these were identified already decades ago. (lu.se)
  • Considering the destructive potential of the complement system, it is no surprise that nearly half of the system's proteins are involved in its inhibition. (lu.se)
  • Some microorganisms either produce a functional mimic of a complement regulatory protein or hijack host's regulatory proteins. (lu.se)
  • Spontaneous or surface-dependent hydrolysis of supplement proteins 3 (C3) to C3(H2O) particularly activates this pathway in individual bloodstream. (gasyblog.com)
  • Supplement aspect H (CFH) dissociates the C3-convertase and in conjunction with aspect I inactivates the C3b proteins [7], [8]. (gasyblog.com)
  • The complement system is a family of plasma proteins that mediates humoral immune surveillance. (ashpublications.org)
  • Activation of the different pathways is mediated by pathway-specific proteins that recognize molecular patterns on pathogens. (ashpublications.org)
  • The membrane attack complex is initiated when the complement protein C5 convertase cleaves C5 into C5a and C5b. (wikipedia.org)
  • Adipocytes synthesize factor D, the limiting component of the alternative complement pathway, which cleaves C3-bound factor B to its active enzymatic form. (medscape.com)
  • C2a in the convertase complex cleaves C3 releasing C3a and C3b. (abcam.com)
  • This complex cleaves C5 leading to the release of C5a and C5b. (abcam.com)
  • This complex is a C3/C5 convertase that cleaves both complement components C3 and C5. (expasy.org)
  • The alternative complement system pathway is spontaneously activated due to the absence of CD55, leading to activation of a C3 convertase that that cleaves C3 into C3a and C3b. (drugbank.com)
  • C3b cleaves C5 into C5a and C5b. (drugbank.com)
  • The classical, lectin, and alternative pathways converge into a final common pathway when C3 convertase (C3 con) cleaves C3 into C3a and C3b. (msdmanuals.com)
  • In all three pathways, a C3-convertase cleaves and activates component C3 , creating C3a and C3b and causing a cascade of further cleavage and activation events. (wikidoc.org)
  • C3(H2O) binds supplement aspect B and interacts with supplement aspect D to create a fluid stage C3-convertase which cleaves C3 in C3a and C3b. (gasyblog.com)
  • Within a positive reviews loop, C3-convertase cleaves C3 and yet another C3b molecule suits C3bBb to create the C5-convertase. (gasyblog.com)
  • Following these cleavage events, complement pathway activation continues as in the classical pathway. (abcam.com)
  • Complement factor H functions as a cofactor in the inactivation of C3b by factor I. It makes C3b susceptible to cleavage by factor I, resulting in iC3b. (bio-rad-antibodies.com)
  • Both provide a path to the cleavage of C3 which is a central event in complement activation. (vetsci.co.uk)
  • C3 cleavage may result in formation of the membrane attack complex (MAC), the cytotoxic component of the complement system. (msdmanuals.com)
  • Three activating pathways comprise the complement cascade (classical, mannose-binding lectin, and alternative), and all converge on the proteolytic cleavage of C3 to generate an arsenal of inflammatory mediators, including the opsonin C3b and the anaphylatoxin C3a. (molvis.org)
  • If left to propagate, the accumulation of C3b triggers cleavage of C5 to produce C5a and C5b, with the latter inducing the assembly of a membrane attack complex (MAC) that binds to cell surfaces, forming transmembrane channels that cause cytolysis or apoptosis of the target cells. (molvis.org)
  • The current dogma of CFH function is to downregulate the complement cascade, by inhibiting cleavage of C3, limiting the generation of byproducts that spur inflammation. (molvis.org)
  • Furthermore, lectin pathway activation by T. cruzi required the MBL-associated serine protease 2 (MASP2) activity resulting in C2 factor cleavage. (herts.ac.uk)
  • Biochemical studies revealed that the CRIT extracellular domain 1 inhibits MASP2 cleavage of C2 factor and thereby impairs C3 convertase formation. (herts.ac.uk)
  • Most inhibitors act on complement convertases through increased dissociation of these enzymatic complexes (acceleration of decay) or through promoting enzymatic cleavage of activated complement factors C3b or C4b by a serine proteinase factor I (FI). (lu.se)
  • BAY 73-4506 C5 cleavage initiates the terminal supplement pathway and mediates irritation aswell as the forming of a cell membrane strike [9]. (gasyblog.com)
  • Activation of the lectin pathway may also trigger the coagulation cascade via MASP-2 cleavage of prothrombin to thrombin. (biomedcentral.com)
  • The cleavage of C5 by this convertase initiates the lytic or terminal pathway. (hycultbiotech.com)
  • In contrast to the activation pathways, which require enzymatic cleavage for activation, the terminal pathway relies on conformational changes induced by binding. (hycultbiotech.com)
  • An activation peptide, C5a, which is an anaphylatoxin that possesses potent spasmogenic and chemotactic activity, is derived from the alpha polypeptide via cleavage with a convertase. (innatedb.com)
  • The C5b macromolecular cleavage product can form a complex with the C6 complement component, and this complex is the basis for formation of the membrane attack complex, which includes additional complement components. (innatedb.com)
  • C3 nephritic factor is a serum immunoglobulin G that interacts with the C3bBb alternative pathway convertase to activate C3. (medscape.com)
  • Adipocytes synthesize C3, factor B, and factor D (adipsin), which allows C3bBb to be formed locally, but which usually does not result in the activation of the terminal lytic part of the complement pathway (C5-9).The IgG antibody, C3Nef, prevents the alternative complement C3-convertase C3Bb from dissociative inactivation, resulting in adipocyte lysis. (medscape.com)
  • Factor H functions as a cofactor in the inactivation of C3b by factor I and also increases the rate of dissociation of the C3bbb complex (C3 convertase) and the (C3b)nbb complex (C5 convertase) in the alternative complement pathway. (lu.se)
  • Properdin can both initiate and positively regulate the alternative pathway activity together with C3 and factors B, D, I and H. It binds to C3b where It stabilizes the labile C3bBb convertase which is deposited on immune complexes or foreign surfaces. (hycultbiotech.com)
  • Thereby enhancing the AP by stimulation of amplification of C3bBb-convertase formation in competition with catabolism of C3b by factor I, which uses factor H as a cofactor. (hycultbiotech.com)
  • Factor H also inhibits the formation of the C3bBb complex (C3 convertase) and increases the rate of dissociation of both C3 convertase and the (C3b)NBB complex (C5 convertase). (bio-rad-antibodies.com)
  • More than 80% of patients with MPGN II are positive for serum C3 nephritic factor (C3NeF), an autoantibody directed against C3bBb, the convertase of the alternative pathway of the complement cascade. (findzebra.com)
  • Properdin stabilizes five to ten-fold both fluid-phase (C3(H2O)Bb) and surface-bound (C3bBb) C3-convertase of the choice pathway [7]. (gasyblog.com)
  • The central functional domains are located in the N terminus (SCR domain 1-4) forming the regulatory domain that controls complement activation at the level of the alternative pathway convertase C3bBb and in the C terminus (SCRs 18-20) that forms the surface binding and attachment region of the protein. (abdominalkey.com)
  • It is therefore possible that inhibiting complement activation would reduce inflammation, lead to reduced fibrosis and preservation of renal function. (wjgnet.com)
  • Compounds comprising peptides and peptidomimetics capable of binding the C3 protein and inhibiting complement activation are disclosed. (justia.com)
  • In particular, this invention provides peptides and peptidomimetics capable of binding the C3 protein and inhibiting complement activation. (justia.com)
  • Advancements in the understanding of the etiopathogenesis of aHUS paved the way for the successful development of anticomplement therapies (complement C5 inhibitors), which have revolutionized the treatment of aHUS. (jrheum.org)
  • Learn about the three pathways lead to complement activation and some of their key inhibitors. (abcam.com)
  • To date, there are no inhibitors of complement activation used in the clinic, though certain candidates for clinical use exist, specifically, a recombinant form of complement receptor 1 known as soluble complement receptor 1 (sCR1) and a humanized monoclonal anti-C5 antibody (5G1.1-scFv). (justia.com)
  • These two latter pathogenic mechanisms are the target of novel strategies of anti-complement treatments, which can be split into terminal and proximal complement inhibitors. (frontiersin.org)
  • Many novel terminal complement inhibitors are now in clinical development: they all target C5 (as eculizumab), potentially paralleling the efficacy and safety profile of eculizumab. (frontiersin.org)
  • Indeed, proximal inhibitors are designed to interfere with early phases of complement activation, eventually preventing C3-mediated extravascular hemolysis in addition to intravascular hemolysis. (frontiersin.org)
  • Indeed, we envision a new scenario of therapeutic complement inhibition, where proximal inhibitors (either anti-C3, anti-FD or anti-FB) may prove effective for the treatment of PNH, either in monotherapy or in combination with anti-C5 agents, eventually leading to drastic improvement of hematological response. (frontiersin.org)
  • Several of these inhibitors circulate in blood whereas others are expressed on virtually all cells of the body to protect self-tissue from complement attack. (lu.se)
  • Several complement inhibitors targeting various complement pathways are in clinical trials for the treatment of HSCT-TMA. (biomedcentral.com)
  • They are produced in response to a growing demand for quantitative investigation of complement inhibitors or regulators at lower sample dilutions. (hycultbiotech.com)
  • Therefore, CVF continuously activates complement, resulting in the depletion of complement activity. (expasy.org)
  • The bound C3b activates the next complement component C5, which again splits up into the subcomponents C5a and C5b upon activation. (vetsci.co.uk)
  • Here, we present the first evidence that T. cruzi activates the complement lectin pathway. (herts.ac.uk)
  • This complex is arranged in nine subunits (six disulfide-linked dimers of A and B, and three disulfide-linked homodimers of C). C1q has binding sites for antibodies (the heavy chain of IMMUNOGLOBULIN G or IMMUNOGLOBULIN M). The interaction of C1q and immunoglobulin activates the two proenzymes COMPLEMENT C1R and COMPLEMENT C1S, thus initiating the cascade of COMPLEMENT ACTIVATION via the CLASSICAL COMPLEMENT PATHWAY. (lookformedical.com)
  • Therefore, mAb 1340 represents a powerful properdin inhibitor ideal for additional research to comprehend the exact systems how properdin activates the match C3-convertase also to determine quantitative degrees of properdin in natural samples. (gasyblog.com)
  • This provokes endotheliopathy, which activates two independent molecular pathways: inflammatory and microthrombotic. (biomedcentral.com)
  • C1 is the first molecule in the classical complement cascade and comprises C1q and two molecules of C1r and C1s respectively. (abcam.com)
  • Activation of the complement cascade, with the formation of the effector MAC unit, results in cytotoxic and cytolytic reactions. (dadamo.com)
  • The main functions of the complement cascade and its role in the acute inflammatory reaction are summarized in Table 1. (dadamo.com)
  • The complement system is a complex, tightly regulated protein cascade involved in pathogen defense and the pathogenesis of several diseases. (bvsalud.org)
  • Here, we fused MAP-2 to the four N-terminal domains of CD55 generating a targeted chimeric inhibitor to modulate complement activation at two different levels of the complement cascade. (bvsalud.org)
  • The complement system is known as a cascade because of the triggering and amplification of further components of the system. (vetsci.co.uk)
  • The whole complement cascade can be triggered in its entirety in a matter of microseconds. (vetsci.co.uk)
  • During the activation process the smaller 'a' subcomponent peptides which are formed, mediate many of the other effects caused by the complement cascade, for example acting as chemoattractants. (vetsci.co.uk)
  • There are three types of complement cascade, the classical and alternative pathways and the Mannan-binding lectin pathway. (vetsci.co.uk)
  • However in the alternative pathway the complement cascade does not require that an antibody binds to the antigen. (vetsci.co.uk)
  • The complement system is an enzyme cascade that helps defend against infection. (msdmanuals.com)
  • Dysregulation of the complement cascade contributes to a variety of retinal dystrophies, including age-related macular degeneration (AMD). (molvis.org)
  • The activation of an immune cascade known as the complement system is a crucial factor in the etiology of age-related macular degeneration (AMD). (molvis.org)
  • This invention relates to activation of the complement cascade in the body. (justia.com)
  • 2006) summarized features of MPGN relevant to the complement cascade. (findzebra.com)
  • This leads to initiation of cascade of enzymatic cleavages and formation of crucial enzymatic complexes (C3 and C5 convertases), release of pro-inflammatory anaphylatoxins (C5a, C3a) that attract white blood cells and finally formation of membrane attack complex (MAC, pore in a membrane). (lu.se)
  • The molecular causes for these defects are often linked to the defective regulation or inappropriate activation of the complement-as well as of the coagulation cascade. (abdominalkey.com)
  • Together, activation of complement and the coagulation cascade lead to a procoagulant state that may result in development of HSCT-TMA. (biomedcentral.com)
  • Cold agglutinin-driven classical pathway activity terminates prior to the initiation of the terminal cascade in CAD patient blood. (ashpublications.org)
  • Complement activation results in an enzymatic cascade leading to the production of opsonins and anaphylatoxins responsible for clearing pathogens and initiating inflammation. (ashpublications.org)
  • The membrane-bound complement inhibitor CD55 acts on the C3/C5 convertase level. (bvsalud.org)
  • aHUS is associated with deregulated complement systems, and is treated with eculizumab, a terminal C5 complement inhibitor [ 1 , 2 ]. (clinmedjournals.org)
  • Pegcetacoplan is a complement inhibitor indicated in the treatment of adults with paroxysmal nocturnal hemoglobinuria. (drugbank.com)
  • Pegcetacoplan is a complement inhibitor indicated in the treatment of paroxysmal nocturnal hemoglobinuria (PNH). (drugbank.com)
  • Pegcetacoplan is a complement C3 inhibitor that prevents complement-mediated hemolysis of red blood cells in patients with paroxysomal nocturnal hemoglobinuria. (drugbank.com)
  • This pathway is regulated by C1 inhibitor (C1-INH). (msdmanuals.com)
  • In addition, we demonstrate that the infectious stage of T. cruzi inhibits the lectin pathway activation and complement killing expressing the complement C2 receptor inhibitor trispanning (CRIT) protein. (herts.ac.uk)
  • C4b-binding protein (C4BP) is the major soluble inhibitor of the classical and lectin pathways whereas factor H (FH) inhibits the alternative route. (lu.se)
  • The complement system is an integral part of the innate immune system but also augments adaptive immune responses. (wjgnet.com)
  • The innate immune system comprises physical, chemical, and biological barriers (e.g., the skin , gastric acid , commensal organisms ) and both cellular (e.g., granulocytes , natural killer cells , mast cells ) and humoral ( complement system ) defense mechanisms . (amboss.com)
  • Now, we know that complement system, protecting the host through innate immune system, could trigger harmful endothelial pathogenesis. (biomedcentral.com)
  • Another complement protein, C6, binds to C5b. (wikipedia.org)
  • The complement system is a tightly regulated, cascading protein network representing a key component linking the innate and humoral immune systems. (jrheum.org)
  • 7 - 9 A TMA is called "primary" when a genetic or acquired defect in a complement protein is identified (as in atypical hemolytic uremic syndrome [aHUS]) or "secondary" when occurring in the context of another disease process or factor such as infection, autoimmune disease, malignancy, or drugs. (jrheum.org)
  • The complement system is a tightly regulated, cascading protein network that performs multiple roles in homeostasis and disease prevention and is a key component of both the innate and the humoral immune systems. (jrheum.org)
  • Three-dimensional structure of a complement control protein module in solution. (lu.se)
  • Positive regulator plasma protein properdin is critical for the alternative pathway of complement. (hycultbiotech.com)
  • In mammals , complement component C3 is the most important protein of the complement system as it is activated by all three pathways (classical, lectin, and alternative pathways), and its various activation products are crucial for mediating, directly or indirectly, virtually all biological functions of complement. (expasy.org)
  • The important protein components of complement are number C1 to C9 (they are numbered in their order of discovery however and not their order of action as you will see later). (vetsci.co.uk)
  • A complement protein attacking an invader. (wikidoc.org)
  • 1987) studied a family in which 3 female sibs had undetectable levels of factor H and C3 nephritic factor, low levels of factor B, C3, and C5 (see 120500), and normal levels of C4-binding protein (120830), factor I (217030), and classic pathway factors. (findzebra.com)
  • Pathogenic variants in complement protein genes were rare. (glomcon.org)
  • The protein encoded by this gene is the fifth component of complement, which plays an important role in inflammatory and cell killing processes. (innatedb.com)
  • 8 , 11 - 14 Numerous stimuli can drive the activation of the complement system, including apoptotic debris, pathogens, and antibody-antigen complexes, in addition to ischemia-reperfusion injuries associated with organ transplantation. (jrheum.org)
  • 4 , 13 Complement plays a crucial role in host defense against foreign bodies by promoting phagocyte-mediated clearance of cell debris through activation of an inflammatory response, opsonization of pathogens, and lysis of susceptible bacteria and cells. (jrheum.org)
  • The complement system is the first line of defense against pathogens and facilitates elimination of apoptotic and damaged cells. (hycultbiotech.com)
  • The complement system helps clear pathogens from an organism. (dadamo.com)
  • The complement system is the first line of immunological defense against foreign pathogens. (justia.com)
  • Kupffer cells and other macrophage cell types help clear complement-coated pathogens. (wikidoc.org)
  • However, only in recent years it has become apparent that complement not only plays a major role in innate defense against pathogens but also identifies foreign materials and removes waste (immune complexes and dying cells). (lu.se)
  • Invading pathogens activate complement either spontaneously due to differences in envelope/membrane composition compared to host (alternative and lectin pathways) or through antibody binding (classical pathway). (lu.se)
  • The pathways that lead to fibrosis are not fully understood, although chronic non-resolving inflammation in the kidney is likely to drive the fibrotic response that occurs. (wjgnet.com)
  • In this review we discuss current evidence that complement activation contributes to progression of CKD, how complement could cause renal inflammation and whether complement inhibition would slow progression of renal disease. (wjgnet.com)
  • Complement activation occurs in progressive chronic kidney disease and may contribute to the chronic inflammation that is characteristically found in the kidney. (wjgnet.com)
  • It is important in regulating the complement pathway, preventing unnecessary inflammation which can damage the host tissue. (bio-rad-antibodies.com)
  • The complement system is a potent mechanism for initiating and amplifying inflammation. (dadamo.com)
  • Pathogenic mechanisms of bradykinin mediated diseases: dysregulation of an innate inflammation pathway. (mastattack.org)
  • These data improve our understanding of the impact of C3 inhibition in subretinal inflammation and inform the development of treatments for targeting complement activation in diseases such as AMD. (molvis.org)
  • Endothelial injury can trigger activation of the complement system, promoting inflammation and the development of endothelial injury syndromes, ultimately leading to organ damage and failure. (biomedcentral.com)
  • Activated inflammatory pathway promotes the release of inflammatory cytokines and triggers inflammation. (biomedcentral.com)
  • Homo sapiens] C5 is the fifth component of complement and is comprised of alpha (C5a) and beta (C5b) polypeptide chains, C5a is a complement anaphylatoxin that can stimulate the generation of nitric oxide along with the secretion of histamine and leukotriene LTC4 from several cell types and can bring about an increase in vascular permeability that facilitates eosinophil accumulation at sites of allergic inflammation. (innatedb.com)
  • The latter binds to the C3 convertase complex to form C4b2a3b, the classical pathway C5 convertase. (abcam.com)
  • describes the use of a phage-displayed combinatorial random peptide library to identify a 27-residue peptide that binds to C3 and inhibits complement activation. (justia.com)
  • The larger fragment C3b binds with C3 convertase to form C5 convertase. (lookformedical.com)
  • Lipodystrophy is often associated with glomerulonephritis, low C3 serum complement levels, and the presence of a C3 nephritic factor. (medscape.com)
  • C3 nephritic factor induces the lysis of adipocytes that secrete adipsin, a product identical to complement factor D. The distribution of the lipoatrophy is postulated to be dictated by the variable amounts of adipsin secreted by the adipocytes at different locations. (medscape.com)
  • Small amounts of C3b are constantly being formed in the circulation, which are inactivated by factors H and I. The binding of C3b to a foreign antigen decreases its affinity for factor H and allows for the formation of increasing amounts of the alternate pathway convertase. (medscape.com)
  • A further complicating factor is the cross-talk between complement, neutrophils, and coagulation pathways in the pathophysiology of TMA. (jrheum.org)
  • Inappropriate activation or dysregulation of the alternative pathway is a critical factor in development of several autoimmune conditions. (hycultbiotech.com)
  • When the alternative pathway is activated either directly or by the classical pathway via the amplification loop, C3b can bind whole Factor B. Factor D can then cleave Factor B in a single site to yield Ba and Bb. (quidel.com)
  • Mouse anti Human Complement Factor H antibody, clone 028B-244.2.10X (10-10) recognizes human complement factor H which exists in 2 forms. (bio-rad-antibodies.com)
  • The second group of receptors reacts with small complement fragments (C4a, C3a, C5a) as well as with C1q, Ba, Bb and factor H. Stimulation of these receptors results in various biological effects (chemotaxis, secretion of vasoactive amines, mediators of the inflammatory and anaphylactic reaction etc. (dadamo.com)
  • Alternative pathway components are often lettered (eg, factor B, factor D) or named (eg, properdin). (msdmanuals.com)
  • Alternate pathway activation occurs when components of microbial cell surfaces (eg, yeast walls, bacterial cell wall lipopolysaccharide [endotoxin]) or immunoglobulin (eg, nephritic factor, aggregated IgA) cleave small amounts of C3. (msdmanuals.com)
  • This pathway is regulated by properdin, factor H, and decay-accelerating factor (CD55). (msdmanuals.com)
  • At the moment there are three strategies of proximal complement inhibition: anti-C3 agents, anti-factor D agents and anti-factor B agents. (frontiersin.org)
  • Complement factor H deficiency (CFHD) can manifest as several different phenotypes, including asymptomatic, recurrent bacterial infections, and renal failure. (findzebra.com)
  • Laboratory features usually include decreased serum levels of factor H , complement component C3 (120700), and a decrease in other alternative pathway components, indicating activation of the alternative complement pathway. (findzebra.com)
  • See also complement factor I deficiency (610984), which shows phenotypic overlap with this disorder. (findzebra.com)
  • In 1 family, of Polish origin, a teenaged male had vasculitis, thrombocytopenia, proteinuria, and depressed levels of serum factor H and complement component C3. (findzebra.com)
  • 1986) reported a consanguineous Algerian family in which 2 brothers had early-onset glomerulonephritis with C3 deposits and low levels (less than 10% of normal) of complement factor H . The factor H deficiency was defined by undetectable complement hemolytic activity by the classic (CH50) and alternate (AP50) pathways, and low levels of C3 and factor B (138470). (findzebra.com)
  • Activated microthrombotic pathway mediates platelet activation and exocytosis of unusually large von Willebrand factor multimers (ULVWF) from ECs and initiates microthrombogenesis. (biomedcentral.com)
  • The local amplification process leads to the creation of the alternative pathway C5 convertase, C3bBb3b, and initiates the terminal pathway of complement activation. (hycultbiotech.com)
  • the classical pathway initiated by antibodies bound to the surface of foreign bodies and the alternative and lectin pathways that provide an antibody-independent mechanism for complement activation, induced by the presence of bacteria and other micro-organisms. (abcam.com)
  • Antibodies depend on complement for many of their biological activities. (vetsci.co.uk)
  • In the early 20th century, this controversy was resolved when it was understood that complement can act in combination with specific antibodies, or on its own in a non-specific way. (wikidoc.org)
  • The classical complement pathway typically requires antibodies for activation (specific immune response), while the alternative and mannose-binding lectin pathways can be activated by C3 hydrolysis or antigens without the presence of antibodies (non-specific immune response). (wikidoc.org)
  • The classical pathway is triggered by activation of the C1-complex (which consists of one molecule C1q and two molecules C1r and C1s), either by C1q's binding to antibodies from classes M and G, complexed with antigens , or by its binding C1q to the surface of the pathogen. (wikidoc.org)
  • In addition, the following are also involved: antimicrobial peptides, natural antibodies, the complement system, NK cells, and gamma delta T lymphocytes [ 3 ] . (encyclopedia.pub)
  • In the case of the CP, activation is driven by immune complexes containing complement-fixing antibodies. (ashpublications.org)
  • The functions of complement include the attraction of inflammatory cells, opsonization to promote phagocytosis, immune complex clearance and direct microbial killing through the formation of the membrane attack complex (MAC). (abcam.com)
  • Thus, the development of complement modulators has risen as a potential treatment for complement-driven inflammatory pathologies. (bvsalud.org)
  • Endothelial injury triggers activation of the complement system-significantly through the lectin pathway-via altered cell-surface patterns on injured endothelial cells, initiating an inflammatory response [ 7 ]. (biomedcentral.com)
  • The formation of TCC causes lysis of cells or can trigger a variety of cellular metabolic pathways resulting in the synthesis and release of inflammatory mediators. (hycultbiotech.com)
  • Homo sapiens] C5 fragments are released from neutrophils upon their activation of the alternative complement pathway and this further amplifies neutrophil pro-inflammatory responses. (innatedb.com)
  • Although complement is one of the first lines of defense against infection, hyperactivity of specific pathways has been described in numerous autoimmune and inflammatory disease settings. (ashpublications.org)
  • Here we discuss evidence for the role of lectin pathway activation in endothelial injury-associated complications of HSCT and how targeting complement activity may provide therapeutic benefit for patients with HSCT-TMA. (biomedcentral.com)
  • This interaction results in the formation of C4b2a, which is the classic pathway C3b convertase. (medscape.com)
  • C2a remains associated with C4b to form the classical pathway C3 convertase (C4b2a). (abcam.com)
  • Quidel's purified human complement components are useful in a wide variety of immunochemical and research applications. (quidel.com)
  • Our group investigates the physiological regulation of human complement system as well as pathologic situations when this regulation fails. (lu.se)
  • Different fragments, released from individual components during complement activation, operate by a non-cytolytic mechanism through specific receptors present on various cell types. (dadamo.com)
  • From the functional standpoint, complement receptors can be divided into two types: the adherent type and the other receptors. (dadamo.com)
  • T. cruzi infectious stages resist complement-mediated killing by expressing surface receptors, which dissociate or prevent C3 convertase formation. (herts.ac.uk)
  • C3 can be cleaved into COMPLEMENT C3A and COMPLEMENT C3B, spontaneously at low level or by C3 CONVERTASE at high level. (lookformedical.com)
  • All three pathways of the complement system (classical, lectin and alternative pathways) initiate the formation of MAC. (wikipedia.org)
  • The normal complement system consists of the classic and alternative pathways. (medscape.com)
  • Therefore, a clearer understanding of the role of the complement system in TMA associated with other conditions will help to identify patients who would benefit from these therapies. (jrheum.org)
  • Activation of the complement system occurs through the classical (CP), lectin (LP), or alternative (AP) pathways. (jrheum.org)
  • The complement system is a heat-labile component of blood that confers bactericidal properties. (abcam.com)
  • The actions of the complement system affect both innate immunity and acquired immunity. (dadamo.com)
  • The complement system influence the activity of numerous cells, tissues and physiological mechanism of the body. (dadamo.com)
  • Anaphylatoxins are proteolytic products of the serine proteases of the complement system: C3a, C4a and C5a. (dadamo.com)
  • The components of the complement system can be found throughout the body in fluids, providing the body with a systemic means of protection. (vetsci.co.uk)
  • The complement system is the first line of defence against pathogen infection and can be activated by the classic, alternative and lectin pathways. (herts.ac.uk)
  • Trypanosoma cruzi, the causative agent of Chagas disease, has to evade complement system killing and invade the host cells to progress in infection. (herts.ac.uk)
  • Our findings establish that the complement lectin pathway recognizes T. cruzi and provide molecular insights into how the infectious stage inhibits this activation to resist complement system killing. (herts.ac.uk)
  • The term "complement" was introduced by Paul Ehrlich in the late 1890s, as part of his larger theory of the immune system. (wikidoc.org)
  • Ehrlich therefore named this heat-labile component "complement", because it is something in the blood which "complements" the cells of the immune system. (wikidoc.org)
  • The underlying pathophysiology of C3 glomerulopathy involves dysregulation and overactivation of the alternative pathway of the complement system. (glomcon.org)
  • The complement system can be an essential area of the innate disease fighting capability by acting as an initial type of defense which is stabilized by properdin, the only real known positive regulator of the choice complement pathway. (gasyblog.com)
  • In this article, we review the role of the complement system in HSCT-TMA pathogenesis, with a focus on the lectin pathway. (biomedcentral.com)
  • Mannan-binding lectin (MBL) and MBL-associated serine proteases (MASPs) are involved in the initial step of the lectin pathway of complement activation. (abcam.com)
  • Serum serine proteases which participate in COMPLEMENT ACTIVATION. (bvsalud.org)
  • This prevents these components of the classical and of alternative complement pathways from forming a positive feedback loop. (bio-rad-antibodies.com)
  • While MAP-2 and CD55 alone showed a minor inhibition of the three complement pathways when co-incubated with serum (IC50MAP-2+CD55 1-4 = 60.98, 36.10, and 97.01 nM on the classical, lectin, and alternative pathways, respectively), MAP-2:CD551-4 demonstrated a potent inhibitory activity (IC50MAP-2:CD55 1-4 = 2.94, 1.76, and 12.86 nM, respectively). (bvsalud.org)
  • In a similar manner to the classical pathway, the alternative pathway works by forming the MAC (membrane attack complex). (vetsci.co.uk)
  • Classical pathway components are labeled with a C and a number (eg, C1, C3), based on the order in which they were identified. (msdmanuals.com)
  • This pathway otherwise resembles the classical pathway structurally and functionally. (msdmanuals.com)
  • Deficiencies of an early component of the classical complement pathway (C1q, C1r, C1s, C2, C4) have been associated with lupus like autoimmune conditions. (mastattack.org)
  • Its activation through the classical, alternative or lectin pathways leads to the generation of anaphylatoxic peptides C3a and C5a and formation of the C5b-9 membrane attack complex. (justia.com)
  • The classical and alternative complement pathways. (wikidoc.org)
  • A glycoprotein that is central in both the classical and the alternative pathway of COMPLEMENT ACTIVATION. (lookformedical.com)
  • We are very proud of our newly developed human classical and alternative complement pathway assays. (hycultbiotech.com)
  • Activation of the classical pathway (CP) of complement is often associated with autoimmune disorders in which disease pathology is linked to the presence of an autoantibody. (ashpublications.org)
  • Complement can be triggered by one of three distinct pathways: the classical pathway (CP), alternative pathway (AP), or lectin (LP) pathway. (ashpublications.org)
  • They are enzymes in the various COMPLEMENT ACTIVATION pathways. (lookformedical.com)
  • The three distinct activation pathways of complement converge with the formation of a C5 convertase. (hycultbiotech.com)
  • Stabilization from the convertases is certainly compared by CFH-mediated dissociation of Bb although immediate inhibitory legislation of properdin by CFH isn't known. (gasyblog.com)
  • The role of complement dysregulation on vascular endothelial cells has been well established in atypical hemolytic uremic syndrome (aHUS), a thrombotic microangiopathy (TMA) characterized by microangiopathic hemolytic anemia, thrombocytopenia, and target organ injury. (jrheum.org)
  • Activation of alternate complement pathway has been demonstrated in most patients. (medscape.com)
  • Activation of an alternate complement pathway, C3 hypocomplementemia with lysis of adipocytes induced by C3NeF, has been implicated. (medscape.com)
  • The classic and alternate pathway convertases cause C3 activation, forming C3a and C3b. (medscape.com)
  • C3b is an opsonin itself, and C3 convertase facilitates the activation of the terminal pathway and the formation of the membrane attack complex C5b-9. (medscape.com)
  • Terminal complement pathway deficiency Paroxysmal nocturnal haemoglobinuria Perforin Pore-forming toxin Xie CB, Jane-Wit D, Pober JS (2020). (wikipedia.org)
  • Mutations in this gene cause complement component 5 deficiency, a disease where patients show a propensity for severe recurrent infections. (innatedb.com)
  • Another form of lipodystrophy that fits the classification of "acquired partial" not involving the complement pathway is associated with hematopoietic stem cell transplantation (HSCT) to treat leukemia or neuroblastoma. (medscape.com)
  • Antibody-mediated complement activation leads to MAC deposition on the surface of infected cells. (wikipedia.org)
  • By inhibiting cold agglutinin-mediated complement deposition on the cellular membrane, TNT003 prevents RBCs from being phagocytosed. (ashpublications.org)
  • Anemia occurs as a result of autoantibody-mediated CP activation on the surface of the erythrocyte, leading to the deposition of complement opsonins that drive extravascular hemolysis in the liver. (ashpublications.org)
  • We collected 40 individual CAD patient samples and showed that TNT003 prevented cold agglutinin-mediated deposition of complement opsonins that promote phagocytosis of RBCs. (ashpublications.org)
  • Monoclonal antibody aE11 reacts with a C9 neoantigen of the terminal complement complex (TCC). (hycultbiotech.com)
  • After binding of C8, a variable number of C9 molecules associate with the C5b678 complex, which is also termed- the terminal complement complex (TCC). (hycultbiotech.com)
  • C3b is able to activate a number of C5 molecules like this. (vetsci.co.uk)
  • The pathogenesis of pregnancy-associated aHUS associated with defects in complement regulatory systems, which may be unmasked during pregnancy, a compliment-amplifying state [ 5 , 7 ]. (clinmedjournals.org)
  • All Quidel complement components are tested for functional activity in a standard lytic or applicable functional assay and for biochemical purity by SDS- polyacrylamide gel electrophoresis. (quidel.com)
  • The MAC is composed of the complement components C5b, C6, C7, C8 and several C9 molecules. (wikipedia.org)
  • Dead cells are also removed by these complement molecules. (mastattack.org)
  • 10 This review aims to provide an assessment of the nature and extent of complement involvement in the underlying pathophysiology of TMA associated with autoimmune diseases that will help to stratify patients for targeted therapy. (jrheum.org)
  • When expanding the autoimmune profile to include "lupus-like" conditions such as those often associated with complement deficiencies, a much higher prevalence of autoantibodies was found in HAE patients. (mastattack.org)
  • Complement-mediated tissue injury has also been found to result from bioincompatibility situations such as those encountered in patients undergoing dialysis or cardiopulmonary bypass. (justia.com)
  • Patients with MPGN type II without C3NeF often have mutations in the CFH gene, which also results in prolonged activation of C3 convertase. (findzebra.com)
  • The physiological relevance of complement is demonstrated by diseases affecting patients lacking complement components: recurrent infections, autoimmune diseases and glomerulonephritis. (lu.se)
  • TCC is present in normal human plasma and increased in patients with complement activation. (hycultbiotech.com)
  • Anti-complement Treatment for Paroxysmal Nocturnal Hemoglobinuria: Time for Proximal Complement Inhibition? (frontiersin.org)
  • However, novel anti-C5 agents do not improve hematological response to eculizumab, even if some seem associated with a lower risk of breakthrough hemolysis caused by pharmacokinetic reasons (it remains unclear whether more effective inhibition of C5 is possible and clinically beneficial). (frontiersin.org)
  • Preliminary data clearly demonstrate that proximal complement inhibition is pharmacologically feasible and apparently safe, and may drastically improve the hematological response to complement inhibition in PNH. (frontiersin.org)
  • The result of cytotoxic complement reaction may be beneficial for the body (elimination of the infectious agent or damaged cells) or harmful (damage to autologous normal cells by immunopathological reactions). (dadamo.com)
  • The classic pathway is activated by the interaction of C1 with an antigen-antibody complex. (medscape.com)
  • Paroxysmal nocturnal hemoglobinuria (PNH) is a rare hematological disorder characterized by complement-mediated intravascular hemolysis, bone marrow failure, and severe thrombophilia ( 1 ). (frontiersin.org)