The phenomenon of antibody-mediated target cell destruction by non-sensitized effector cells. The identity of the target cell varies, but it must possess surface IMMUNOGLOBULIN G whose Fc portion is intact. The effector cell is a "killer" cell possessing Fc receptors. It may be a lymphocyte lacking conventional B- or T-cell markers, or a monocyte, macrophage, or polynuclear leukocyte, depending on the identity of the target cell. The reaction is complement-independent.
Enhancement of viral infectivity caused by non-neutralizing antibodies. There are at least two mechanisms known to account for this: mediation by Fc receptors (RECEPTORS, FC) or by complement receptors (RECEPTORS, COMPLEMENT). Either the virus is complexed with antiviral IMMUNOGLOBULIN G and binds to Fc receptors, or virus is coated with antiviral IMMUNOGLOBULIN M and binds to complement receptors.
The phenomenon of target cell destruction by immunologically active effector cells. It may be brought about directly by sensitized T-lymphocytes or by lymphoid or myeloid "killer" cells, or it may be mediated by cytotoxic antibody, cytotoxic factor released by lymphoid cells, or complement.
Bone marrow-derived lymphocytes that possess cytotoxic properties, classically directed against transformed and virus-infected cells. Unlike T CELLS; and B CELLS; NK CELLS are not antigen specific. The cytotoxicity of natural killer cells is determined by the collective signaling of an array of inhibitory and stimulatory CELL SURFACE RECEPTORS. A subset of T-LYMPHOCYTES referred to as NATURAL KILLER T CELLS shares some of the properties of this cell type.
Antibodies produced by a single clone of cells.
The major immunoglobulin isotype class in normal human serum. There are several isotype subclasses of IgG, for example, IgG1, IgG2A, and IgG2B.
Immunoglobulin molecules having a specific amino acid sequence by virtue of which they interact only with the ANTIGEN (or a very similar shape) that induced their synthesis in cells of the lymphoid series (especially PLASMA CELLS).
The property of antibodies which enables them to react with some ANTIGENIC DETERMINANTS and not with others. Specificity is dependent on chemical composition, physical forces, and molecular structure at the binding site.
The demonstration of the cytotoxic effect on a target cell of a lymphocyte, a mediator released by a sensitized lymphocyte, an antibody, or complement.
Immunoglobulins produced in response to VIRAL ANTIGENS.
Immunoglobulins produced in a response to BACTERIAL ANTIGENS.
Established cell cultures that have the potential to propagate indefinitely.
The span of viability of a cell characterized by the capacity to perform certain functions such as metabolism, growth, reproduction, some form of responsiveness, and adaptability.
The production of ANTIBODIES by proliferating and differentiated B-LYMPHOCYTES under stimulation by ANTIGENS.
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.
Antibodies that reduce or abolish some biological activity of a soluble antigen or infectious agent, usually a virus.
A cell line derived from cultured tumor cells.
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.
Test for tissue antigen using either a direct method, by conjugation of antibody with fluorescent dye (FLUORESCENT ANTIBODY TECHNIQUE, DIRECT) or an indirect method, by formation of antigen-antibody complex which is then labeled with fluorescein-conjugated anti-immunoglobulin antibody (FLUORESCENT ANTIBODY TECHNIQUE, INDIRECT). The tissue is then examined by fluorescence microscopy.
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.
Cells grown in vitro from neoplastic tissue. If they can be established as a TUMOR CELL LINE, they can be propagated in cell culture indefinitely.
One of the mechanisms by which CELL DEATH occurs (compare with NECROSIS and AUTOPHAGOCYTOSIS). Apoptosis is the mechanism responsible for the physiological deletion of cells and appears to be intrinsically programmed. It is characterized by distinctive morphologic changes in the nucleus and cytoplasm, chromatin cleavage at regularly spaced sites, and the endonucleolytic cleavage of genomic DNA; (DNA FRAGMENTATION); at internucleosomal sites. This mode of cell death serves as a balance to mitosis in regulating the size of animal tissues and in mediating pathologic processes associated with tumor growth.
A multilineage cell growth factor secreted by LYMPHOCYTES; EPITHELIAL CELLS; and ASTROCYTES which stimulates clonal proliferation and differentiation of various types of blood and tissue cells.
A calcium-dependent pore-forming protein synthesized in cytolytic LYMPHOCYTES and sequestered in secretory granules. Upon immunological reaction between a cytolytic lymphocyte and a target cell, perforin is released at the plasma membrane and polymerizes into transmembrane tubules (forming pores) which lead to death of a target cell.
A measure of the binding strength between antibody and a simple hapten or antigen determinant. It depends on the closeness of stereochemical fit between antibody combining sites and antigen determinants, on the size of the area of contact between them, and on the distribution of charged and hydrophobic groups. It includes the concept of "avidity," which refers to the strength of the antigen-antibody bond after formation of reversible complexes.
Technique using an instrument system for making, processing, and displaying one or more measurements on individual cells obtained from a cell suspension. Cells are usually stained with one or more fluorescent dyes specific to cell components of interest, e.g., DNA, and fluorescence of each cell is measured as it rapidly transverses the excitation beam (laser or mercury arc lamp). Fluorescence provides a quantitative measure of various biochemical and biophysical properties of the cell, as well as a basis for cell sorting. Other measurable optical parameters include light absorption and light scattering, the latter being applicable to the measurement of cell size, shape, density, granularity, and stain uptake.
A soluble substance elaborated by antigen- or mitogen-stimulated T-LYMPHOCYTES which induces DNA synthesis in naive lymphocytes.
The intracellular transfer of information (biological activation/inhibition) through a signal pathway. In each signal transduction system, an activation/inhibition signal from a biologically active molecule (hormone, neurotransmitter) is mediated via the coupling of a receptor/enzyme to a second messenger system or to an ion channel. Signal transduction plays an important role in activating cellular functions, cell differentiation, and cell proliferation. Examples of signal transduction systems are the GAMMA-AMINOBUTYRIC ACID-postsynaptic receptor-calcium ion channel system, the receptor-mediated T-cell activation pathway, and the receptor-mediated activation of phospholipases. Those coupled to membrane depolarization or intracellular release of calcium include the receptor-mediated activation of cytotoxic functions in granulocytes and the synaptic potentiation of protein kinase activation. Some signal transduction pathways may be part of larger signal transduction pathways; for example, protein kinase activation is part of the platelet activation signal pathway.
Cell surface molecules on cells of the immune system that specifically bind surface molecules or messenger molecules and trigger changes in the behavior of cells. Although these receptors were first identified in the immune system, many have important functions elsewhere.
The order of amino acids as they occur in a polypeptide chain. This is referred to as the primary structure of proteins. It is of fundamental importance in determining PROTEIN CONFORMATION.
A 46-kD stimulatory receptor found on resting and activated NATURAL KILLER CELLS. It has specificity for VIRAL HEMAGGLUTININS that are expressed on infected cells.
Morphologic alteration of small B LYMPHOCYTES or T LYMPHOCYTES in culture into large blast-like cells able to synthesize DNA and RNA and to divide mitotically. It is induced by INTERLEUKINS; MITOGENS such as PHYTOHEMAGGLUTININS, and by specific ANTIGENS. It may also occur in vivo as in GRAFT REJECTION.
An activating NK cell lectin-like receptor subfamily that regulates immune responses to INFECTION and NEOPLASMS. Members of this subfamily generally occur as homodimers.
Receptors that are specifically found on the surface of NATURAL KILLER CELLS. They play an important role in regulating the cellular component of INNATE IMMUNITY.
The termination of the cell's ability to carry out vital functions such as metabolism, growth, reproduction, responsiveness, and adaptability.
Proteins prepared by recombinant DNA technology.
Antibodies which react with the individual structural determinants (idiotopes) on the variable region of other antibodies.
Local surface sites on antibodies which react with antigen determinant sites on antigens (EPITOPES.) They are formed from parts of the variable regions of FAB FRAGMENTS.
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.
A 30 kDa stimulatory receptor found on resting and activated NATURAL KILLER CELLS.
Antibodies, often monoclonal, in which the two antigen-binding sites are specific for separate ANTIGENIC DETERMINANTS. They are artificial antibodies produced by chemical crosslinking, fusion of HYBRIDOMA cells, or by molecular genetic techniques. They function as the main mediators of targeted cellular cytotoxicity and have been shown to be efficient in the targeting of drugs, toxins, radiolabeled haptens, and effector cells to diseased tissue, primarily tumors.
An ERYTHROLEUKEMIA cell line derived from a CHRONIC MYELOID LEUKEMIA patient in BLAST CRISIS.
The fission of a CELL. It includes CYTOKINESIS, when the CYTOPLASM of a cell is divided, and CELL NUCLEUS DIVISION.
Lymphocytes responsible for cell-mediated immunity. Two types have been identified - cytotoxic (T-LYMPHOCYTES, CYTOTOXIC) and helper T-lymphocytes (T-LYMPHOCYTES, HELPER-INDUCER). They are formed when lymphocytes circulate through the THYMUS GLAND and differentiate to thymocytes. When exposed to an antigen, they divide rapidly and produce large numbers of new T cells sensitized to that antigen.
A family of serine endopeptidases found in the SECRETORY GRANULES of LEUKOCYTES such as CYTOTOXIC T-LYMPHOCYTES and NATURAL KILLER CELLS. When secreted into the intercellular space granzymes act to eliminate transformed and virus-infected host cells.
Specific molecular sites on the surface of various cells, including B-lymphocytes and macrophages, that combine with IMMUNOGLOBULIN Gs. Three subclasses exist: Fc gamma RI (the CD64 antigen, a low affinity receptor), Fc gamma RII (the CD32 antigen, a high affinity receptor), and Fc gamma RIII (the CD16 antigen, a low affinity receptor).
Glycoproteins found on the membrane or surface of cells.
Proteins secreted from an organism which form membrane-spanning pores in target cells to destroy them. This is in contrast to PORINS and MEMBRANE TRANSPORT PROTEINS that function within the synthesizing organism and COMPLEMENT immune proteins. These pore forming cytotoxic proteins are a form of primitive cellular defense which are also found in human LYMPHOCYTES.
White blood cells formed in the body's lymphoid tissue. The nucleus is round or ovoid with coarse, irregularly clumped chromatin while the cytoplasm is typically pale blue with azurophilic (if any) granules. Most lymphocytes can be classified as either T or B (with subpopulations of each), or NATURAL KILLER CELLS.
Sites on an antigen that interact with specific antibodies.
Immunoglobulins induced by antigens specific for tumors other than the normally occurring HISTOCOMPATIBILITY ANTIGENS.
The 140 kDa isoform of NCAM (neural cell adhesion molecule) containing a transmembrane domain and short cytoplasmic tail. It is expressed by all lymphocytes mediating non-MHC restricted cytotoxicity and is present on some neural tissues and tumors.
The major interferon produced by mitogenically or antigenically stimulated LYMPHOCYTES. It is structurally different from TYPE I INTERFERON and its major activity is immunoregulation. It has been implicated in the expression of CLASS II HISTOCOMPATIBILITY ANTIGENS in cells that do not normally produce them, leading to AUTOIMMUNE DISEASES.

Early membrane rupture events during neutrophil-mediated antibody-dependent tumor cell cytolysis. (1/1442)

Although cell-mediated cytolysis is a fundamental immune effector response, its mechanism remains poorly understood at the cellular level. In this report, we image for the first time transient ruptures, as inferred by cytoplasmic marker release, in tumor cell membranes during Ab-dependent cellular cytolysis. The cytosol of IgG-opsonized YAC tumor cells was labeled with tetra-methylrhodamine diacetate followed by the formation of tumor cell-neutrophil conjugates. We hypothesized that tumor cell cytolysis proceeds via a series of discrete membrane rupture/resealing events that contribute to marker release. To test this hypothesis, we occluded the fluorescence image of the labeled tumor cells by passing an opaque disk into a field-conjugated plane between the light source and the sample. Multiple small bursts of fluorescent label release from tumor cells could be detected using a photomultiplier tube. Similarly, multiple fluorescent plumes were observed at various sites around the perimeter of a target. These findings support a multihit model of target cytolysis and suggest that cytolytic release is not focused at specific sites. Cytolytic bursts were generally observed at 20-s intervals, which match the previously described reduced nicotinamide-adenine dinucleotide phosphate and superoxide release oscillation periods for neutrophils; we speculate that metabolic oscillations of the effector cell drive the membrane damage of the target.  (+info)

Cytotoxicity of human and baboon mononuclear phagocytes against schistosomula in vitro: induction by immune complexes containing IgE and Schistosoma mansoni antigens. (2/1442)

Normal human blood monocytes, pre-incubated at 37 degrees C with sera from patients infected with Schistosoma mansoni, strongly adhered to S. mansoni schistosomula in vitro, whereas no significant adherence was induced by sera from uninfected individuals. Comparable adherence occurred with normal baboon blood monocytes or peritoneal macrophages when these cells were incubated with sera from S. mansoni-infected baboons. Adherence of macrophages to schistosomula was associated with damage to the larvae, as estimated by a 51Cr release technique. Neither adherence nor cytotoxicity was induced by pre-incubation of the schistosomula, instead of the monocytes, with immune serum. The relevant factor in immune serum was heat-labile, but was not a complement component. Absorption and ultracentrifugation experiments showed that immune complexes, containing S. mansoni-specific IgE antibody and soluble parasite antigens, produced monocyte or macrophage adherence and cytotoxicity. Similar observations have been reported previously in the rat model. Since the production of large amounts of IgE is a predominant feature of schistosome infections in man and experimental animals, it is possible that this new mode of mononuclear phagocyte activation could act as an immune effector mechanism against S. mansoni.  (+info)

Improving the efficacy of antibody-interleukin 2 fusion proteins by reducing their interaction with Fc receptors. (3/1442)

Fusion proteins between whole antibodies (Abs) and cytokines (immunocytokines) such as interleukin 2 have shown efficacy in several mouse tumor models despite a circulating half-life that is significantly shorter than that of the original Ab. We have examined the potential mechanisms responsible for clearance and shown that an important factor is enhanced binding to Fc receptor (FcR). Improvements in the half-lives of two different immunocytokines were made by changing the isotype of the human heavy chain C region from IgG1 or IgG3 to those with reduced binding to FcR, e.g., IgG4. The same effect could also be achieved through site-directed mutagenesis of the FcR binding site in the IgG1 H chain. In vitro studies using mouse J774 FcR-expressing cells showed increased binding of interleukin 2-based immunocytokines, relative to their corresponding Abs, and that this was reversed in those fusion proteins made with IgG4 or mutated IgG1 H chains. All of the fusion proteins showing reduced FcR binding also had reduced Ab-dependent cellular cytotoxicity activity, as measured in 4-h chromium release assays. A complete loss of complement-dependent cytotoxicity activity was seen with an IgG4-based immunocytokine derived from an IgG1 Ab with potent activity. Despite these reduced effector functions, the IgG4-based immunocytokines with extended circulating half-lives showed equivalent (in the case of severe combined immunodeficiency mouse xenograft models) or better (in the case of syngeneic models) efficacy in mouse tumor models than the original IgG1-based molecules. These novel immunocytokines may show improved efficacy in therapeutic situations where T cell- rather than natural killer- or complement-mediated antitumor mechanisms are involved.  (+info)

Monoclonal Lym-1 antibody-dependent cytolysis by neutrophils exposed to granulocyte-macrophage colony-stimulating factor: intervention of FcgammaRII (CD32), CD11b-CD18 integrins, and CD66b glycoproteins. (4/1442)

Murine monoclonal antibody (MoAb) Lym-1 is an IgG2a able to bind HLA-DR variants on malignant B cells and suitable for serotherapeutic approaches in B-lymphoma patients. We have previously shown that Lym-1 can synergize with granulocyte-macrophage colony-stimulating factor (GM-CSF) to trigger neutrophil cytolysis towards Raji cells used as a model of B-lymphoma targets. Here we provide evidence for the intervention of certain neutrophil receptors or surface molecules in this model of cell-mediated lysis. The lysis was completely inhibited by the anti-FcgammaRII MoAb IV.3 and unaffected by the anti-FcgammaRIII MoAb 3G8. This suggests that neutrophil cytolysis involves FcgammaRII without cooperation of this receptor with FcgammaRIII. Moreover, the lysis was inhibited by an anti-CD18 MoAb (MEM48) and by a MoAb specific for carcinoembryonic antigen (CEA)-like and glycophosphatidyl inositol (GPI)-linked glycoproteins (CD66b). Using an immunofluorescence staining procedure, cross-linking of CD66b induced the redistribution of CD11b on neutrophils with distinct areas of CD11b clustering via a process susceptible of inhibition by D-mannose. This is consistent with the ability of CD11b-CD18 and CD66b to undergo lectin-like physical interactions on the neutrophil surface. Such a type of interaction is presumably instrumental for neutrophil cytolytic activity in that the lysis was inhibited by D-mannose and enhanced by the MoAb VIM-12, which mimics the cooperation between CD11b and GPI-anchored molecules by specifically interacting with CD11b lectin-like sites. Therefore, the present results prove the absolute requirement for FcgammaRII in neutrophil GM-CSF/Lym-1-mediated cytolysis and, on the other hand, define the crucial role of CD66b and CD11b/CD18 in the expression of the cell lytic potential.  (+info)

Natural cytotoxic and antibody-dependent cellular cytotoxic activity of cells in the decidua basales and metrial glands of pseudopregnant rats with deciduomata. (5/1442)

Cytotoxic cells are present in the uterine wall of pregnant rats. To determine if the cytotoxic activity arises in response to semen or the products of conception, the profile of cytotoxic activity in deciduomata of pseudopregnant rats was examined. To examine NK activity, Yac-1 cells were used as targets in chromium release cytotoxicity assays and an antibody to Yac-1 cells was included in some assays to determine antibody-dependent cellular cytotoxic (ADCC) activity. Cells from the metrial glands and deciduae of deciduomata of rats at days 10 and 13 of pseudopregnancy did not show NK activity but ADCC activity was present. To examine natural cytotoxic (NC) activity, Wehi 164 cells were used as targets in chromium release cytotoxicity assays. Cells isolated from the metrial glands and deciduae of rats at day 10 of pseudopregnancy were able to kill Wehi 164 cells after 21 h assays, thus demonstrating NC activity. The profile of cytotoxic activity in the uterine wall of pseudopregnant rats with deciduomata is similar to that found in pregnancy and is thus independent of semen or the products of conception.  (+info)

Human CD16 as a lysis receptor mediating direct natural killer cell cytotoxicity. (6/1442)

In addition to their role in peptide antigen presentation, class I MHC proteins also play a critical role in inhibiting natural killer (NK) cytotoxicity through interaction with NK inhibitory receptors. Thus, NK cells are cytotoxic to virus-infected and tumor cells that have lost class I MHC protein expression. However, the nature of the receptors involved in the triggering of lysis of target cells is poorly understood. CD16 (Fcgamma receptor III) has been described as a receptor expressed on NK cells that facilitates antibody-dependent cellular cytotoxicity (ADCC) by binding to the Fc portion of various antibodies. However, we show here that CD16 has a broader function and is directly involved in the lysis of some virus-infected cells and tumor cells, independent of antibody binding. The presence of a putative CD16 ligand on appropriate target cells has also been demonstrated by the use of a CD16-Ig fusion protein.  (+info)

C-myc antisense oligodeoxynucleotides can induce apoptosis and down-regulate Fas expression in rheumatoid synoviocytes. (7/1442)

OBJECTIVE: To investigate the role of c-myc in the pathogenesis of rheumatoid arthritis (RA) and the mechanism of synovial apoptosis. METHODS: Using cultured human synoviocytes from patients with RA and c-myc antisense oligodeoxynucleotides (AS ODN), we examined the inhibition of cell proliferation by the MTT assay and the induction of apoptosis with TUNEL staining and fluorescence microscopy. In addition, the effect of c-myc on down-regulation of Fas expression was analyzed by flow cytometry, cytotoxicity assay, and reverse transcriptase-polymerase chain reaction. RESULTS: Treatment with c-myc AS ODN induced inhibition of cell proliferation, along with down-regulation of c-Myc protein and c-myc messenger RNA (mRNA) expression. The morphologic changes of synovial cell death were typical of apoptosis. In addition, c-myc AS ODN treatment down-regulated expression of Fas mRNA but not Fas antigen. Analysis of the involvement of the caspase cascade revealed that the cytotoxic activity of c-myc AS ODN was completely blocked by inhibitors of both caspase 1 (YVAD-FMK) and caspase 3 (DEVD-FMK). CONCLUSION: Our results strongly suggest that c-myc AS ODN might be a useful therapeutic tool in RA and clarify that cell death by c-myc AS ODN is induced through the caspase cascade, similar to Fas-induced apoptosis. In addition, combination therapy with anti-Fas antibody and c-myc AS ODN reduced Fas-dependent cytotoxicity.  (+info)

Differential involvement of the CD95 (Fas/APO-1) receptor/ligand system on apoptosis induced by the wild-type p53 gene transfer in human cancer cells. (8/1442)

The CD95 (Fas/APO-1) system regulates a number of physiological and pathological processes of cell death. The ligand for CD95 induces apoptosis in sensitive target cells by interacting with a transmembrane cell surface CD95 receptor. We previously reported that the recombinant adenovirus-mediated transfer of the wild-type p53 gene caused apoptotic cell death in a variety of human cancer cells. To better understand the mechanism responsible for this cell death signaling, we have investigated the potential involvement of the CD95 receptor/ligand system in p53-mediated apoptosis. The transient expression of the wild-type p53 gene upregulated the CD95 ligand mRNA as well as protein expression in H1299 human lung cancer cells deficient for p53 and in DLD-1 and SW620 human colon cancer cells with mutated p53, all of which constitutively expressed CD95 receptor as shown by a flow cytometric analysis, and induced rapid apoptotic cell death as early as 24 h after gene transfer. However, the sensitivity to the cytolytic effect of agonistic anti-CD95 antibody (CH11) varied among these cell lines: CH11 induced apoptosis in H1299 cells, but not in DLD-1 and SW620 cells despite their abundant CD95 receptor expression, suggesting that the CD95 receptors on DLD-1 and SW620 cells might be inactivated. In addition, an antagonistic anti-CD95 ligand antibody (4H9) that interfered with the CD95-receptor-ligand interaction partially reduced the apoptosis induced by the wild-type p53 gene transfer in H1299 cells, whereas apoptosis of DLD-1 and SW620 cells occurred in the presence of 4H9. Taken together, these findings led us to conclude that the CD95 receptor/ligand system is differentially involved in p53-mediated apoptosis, suggesting that the restoration of the wild-type p53 function may mediate apoptosis through CD95 receptor/ligand interactions as well as an alternative pathway.  (+info)

Antibody-Dependent Cell Cytotoxicity (ADCC) is a type of immune response in which the effector cells of the immune system, such as natural killer (NK) cells, cytotoxic T-cells or macrophages, recognize and destroy virus-infected or cancer cells that are coated with antibodies.

In this process, an antibody produced by B-cells binds specifically to an antigen on the surface of a target cell. The other end of the antibody then interacts with Fc receptors found on the surface of effector cells. This interaction triggers the effector cells to release cytotoxic substances, such as perforins and granzymes, which create pores in the target cell membrane and induce apoptosis (programmed cell death).

ADCC plays an important role in the immune defense against viral infections and cancer. It is also a mechanism of action for some monoclonal antibody therapies used in cancer treatment.

Antibody-Dependent Enhancement (ADE) is a phenomenon in which the presence of antibodies against a particular virus actually enhances the ability of the virus to infect and replicate within host cells, leading to increased severity of infection. This occurs when the antibodies bind to the virus but do not neutralize it, instead facilitating uptake of the virus into immune cells expressing Fc receptors, such as macrophages. The virus can then use these cells as a site for replication and evasion of the host's immune response. ADE has been observed in various viral infections, including dengue fever and respiratory syncytial virus (RSV) infection. It is a concern in the development of vaccines against these viruses, as non-neutralizing antibodies induced by vaccination could potentially enhance subsequent infection with a heterologous strain of the virus.

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.

Natural Killer (NK) cells are a type of lymphocyte, which are large granular innate immune cells that play a crucial role in the host's defense against viral infections and malignant transformations. They do not require prior sensitization to target and destroy abnormal cells, such as virus-infected cells or tumor cells. NK cells recognize their targets through an array of germline-encoded activating and inhibitory receptors that detect the alterations in the cell surface molecules of potential targets. Upon activation, NK cells release cytotoxic granules containing perforins and granzymes to induce target cell apoptosis, and they also produce a variety of cytokines and chemokines to modulate immune responses. Overall, natural killer cells serve as a critical component of the innate immune system, providing rapid and effective responses against infected or malignant cells.

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.

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.

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.

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.

Cytotoxicity tests, immunologic are a group of laboratory assays used to measure the immune-mediated damage or destruction (cytotoxicity) of cells. These tests are often used in medical research and clinical settings to evaluate the potential toxicity of drugs, biological agents, or environmental factors on specific types of cells.

Immunologic cytotoxicity tests typically involve the use of immune effector cells, such as cytotoxic T lymphocytes (CTLs) or natural killer (NK) cells, which can recognize and kill target cells that express specific antigens on their surface. The tests may also involve the use of antibodies or other immune molecules that can bind to target cells and trigger complement-mediated cytotoxicity.

There are several types of immunologic cytotoxicity tests, including:

1. Cytotoxic T lymphocyte (CTL) assays: These tests measure the ability of CTLs to recognize and kill target cells that express specific antigens. The test involves incubating target cells with CTLs and then measuring the amount of cell death or damage.
2. Natural killer (NK) cell assays: These tests measure the ability of NK cells to recognize and kill target cells that lack self-antigens or express stress-induced antigens. The test involves incubating target cells with NK cells and then measuring the amount of cell death or damage.
3. Antibody-dependent cellular cytotoxicity (ADCC) assays: These tests measure the ability of antibodies to bind to target cells and recruit immune effector cells, such as NK cells or macrophages, to mediate cell lysis. The test involves incubating target cells with antibodies and then measuring the amount of cell death or damage.
4. Complement-dependent cytotoxicity (CDC) assays: These tests measure the ability of complement proteins to bind to target cells and form a membrane attack complex that leads to cell lysis. The test involves incubating target cells with complement proteins and then measuring the amount of cell death or damage.

Immunologic cytotoxicity tests are important tools in immunology, cancer research, and drug development. They can help researchers understand how immune cells recognize and kill infected or damaged cells, as well as how to develop new therapies that enhance or inhibit these processes.

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

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

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

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

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.

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.

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

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

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

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.

"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.

Neutralizing antibodies are a type of antibody that defends against pathogens such as viruses or bacteria by neutralizing their ability to infect cells. They do this by binding to specific regions on the surface proteins of the pathogen, preventing it from attaching to and entering host cells. This renders the pathogen ineffective and helps to prevent or reduce the severity of infection. Neutralizing antibodies can be produced naturally in response to an infection or vaccination, or they can be generated artificially for therapeutic purposes.

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.

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.

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.

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.

'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.

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).

Interleukin-3 (IL-3) is a type of cytokine, which is a small signaling protein that modulates the immune response, cell growth, and differentiation. IL-3 is primarily produced by activated T cells and mast cells. It plays an essential role in the survival, proliferation, and differentiation of hematopoietic stem cells, which give rise to all blood cell types. Specifically, IL-3 supports the development of myeloid lineage cells, including basophils, eosinophils, mast cells, megakaryocytes, and erythroid progenitors.

IL-3 binds to its receptor, the interleukin-3 receptor (IL-3R), which consists of two subunits: CD123 (the alpha chain) and CD131 (the beta chain). The binding of IL-3 to its receptor triggers a signaling cascade within the cell that ultimately leads to changes in gene expression, promoting cell growth and differentiation. Dysregulation of IL-3 production or signaling has been implicated in several hematological disorders, such as leukemia and myelodysplastic syndromes.

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

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

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

Antibody affinity refers to the strength and specificity of the interaction between an antibody and its corresponding antigen at a molecular level. It is a measure of how strongly and selectively an antibody binds to its target antigen. A higher affinity indicates a more stable and specific binding, while a lower affinity suggests weaker and less specific interactions. Affinity is typically measured in terms of the dissociation constant (Kd), which describes the concentration of antigen needed to achieve half-maximal binding to an antibody. Generally, a smaller Kd value corresponds to a higher affinity, indicating a tighter and more selective bond. This parameter is crucial in the development of diagnostic and therapeutic applications, such as immunoassays and targeted therapies, where high-affinity antibodies are preferred for improved sensitivity and specificity.

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.

Interleukin-2 (IL-2) is a type of cytokine, which are signaling molecules that mediate and regulate immunity, inflammation, and hematopoiesis. Specifically, IL-2 is a growth factor for T cells, a type of white blood cell that plays a central role in the immune response. It is primarily produced by CD4+ T cells (also known as T helper cells) and stimulates the proliferation and differentiation of activated T cells, including effector T cells and regulatory T cells. IL-2 also has roles in the activation and function of other immune cells, such as B cells, natural killer cells, and dendritic cells. Dysregulation of IL-2 production or signaling can contribute to various pathological conditions, including autoimmune diseases, chronic infections, and cancer.

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.

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

There are several types of immunologic receptors, including:

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

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

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

Natural Cytotoxicity Triggering Receptor 1 (NKp46, also known as NCR1) is a type of receptor found on the surface of natural killer (NK) cells and some T-cells. It is a member of the Natural Cytotoxicity Receptors (NCR) family which are involved in the recognition and elimination of target cells, such as virus-infected or tumor cells, by NK cells.

NKp46 recognizes specific structures on the surface of target cells, known as ligands, and when it binds to these ligands, it triggers a series of events that can lead to the killing of the target cell. The activation of NKp46 has been shown to play an important role in the immune response against viral infections and tumors.

It's worth noting that mutations in the NKp46 gene have been associated with certain immunodeficiency disorders, highlighting its importance in the immune system.

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

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

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

'NK cell lectin-like receptor subfamily K' refers to a group of genes that encode for proteins found on natural killer (NK) cells, which are a type of immune cell. These proteins are known as lectin-like receptors because they bind to carbohydrates in a manner similar to lectins.

The NK cell lectin-like receptor subfamily K includes several different genes, including KLRK1 (which encodes for the protein NKG2D), KLRC1 (which encodes for the protein NKG2A), and KLRD1 (which encodes for the protein CD94). These proteins play important roles in regulating NK cell function, including activating or inhibiting NK cells in response to signals from other cells.

NKG2D, for example, binds to ligands expressed on stressed or infected cells, triggering NK cell activation and killing of those cells. NKG2A, on the other hand, binds to a different set of ligands that can inhibit NK cell activation and help prevent the destruction of healthy cells.

Overall, the NK cell lectin-like receptor subfamily K is an important component of the immune system, helping to regulate NK cell function and protect against infection and cancer.

Natural Killer (NK) cell receptors are a type of cell surface receptors expressed by natural killer cells, which are a crucial component of the innate immune system. These receptors play an essential role in the recognition and elimination of abnormal cells, such as virus-infected or malignantly transformed cells.

There are two major types of NK cell receptors: activating receptors and inhibitory receptors. Activating receptors bind to ligands on the surface of target cells, triggering a signaling cascade that leads to the cytotoxic killing of the abnormal cell. In contrast, inhibitory receptors recognize major histocompatibility complex (MHC) class I molecules on healthy cells and transmit an inhibitory signal, preventing NK cells from attacking normal cells.

The balance between activating and inhibitory signals received by NK cells determines their response to target cells. When the activating signals outweigh the inhibitory ones, NK cells become activated and initiate cytotoxic responses or release cytokines to help coordinate the immune response. Dysregulation of NK cell receptors has been implicated in various diseases, including cancer and autoimmune disorders.

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

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

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

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

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

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.

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

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

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

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

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.

Natural Cytotoxicity Triggering Receptor 3 (NKp30 or NCR3) is a type II transmembrane protein that belongs to the natural cytotoxicity receptors (NCRs) family. It is primarily expressed on natural killer (NK) cells and some T-cell subsets, including CD8+ αβ T cells, CD4+ αβ T cells, and γδ T cells.

NKp30 plays a crucial role in the cytotoxic function of NK cells by mediating their natural cytotoxicity against virus-infected or malignantly transformed cells. It recognizes various ligands present on the surface of target cells, including B7-H6, BAG6, and viral hemagglutinins, leading to the activation of NK cells and subsequent killing of the target cells.

Additionally, NKp30 has been implicated in the regulation of adaptive immune responses by modulating dendritic cell maturation and cytokine production. Dysregulation of NKp30 expression or function has been associated with several pathological conditions, including cancer, viral infections, and autoimmune diseases.

Bispecific antibodies are a type of artificial protein that have been engineered to recognize and bind to two different antigens simultaneously. They are created by combining two separate antibody molecules, each with a unique binding site, into a single entity. This allows the bispecific antibody to link two cells or proteins together, bringing them into close proximity and facilitating various biological processes.

In the context of medicine and immunotherapy, bispecific antibodies are being investigated as a potential treatment for cancer and other diseases. For example, a bispecific antibody can be designed to recognize a specific tumor-associated antigen on the surface of cancer cells, while also binding to a component of the immune system, such as a T cell. This brings the T cell into close contact with the cancer cell, activating the immune system and triggering an immune response against the tumor.

Bispecific antibodies have several potential advantages over traditional monoclonal antibodies, which only recognize a single antigen. By targeting two different epitopes or antigens, bispecific antibodies can increase the specificity and affinity of the interaction, reducing off-target effects and improving therapeutic efficacy. Additionally, bispecific antibodies can bring together multiple components of the immune system, amplifying the immune response and enhancing the destruction of cancer cells.

Overall, bispecific antibodies represent a promising new class of therapeutics that have the potential to revolutionize the treatment of cancer and other diseases. However, further research is needed to fully understand their mechanisms of action and optimize their clinical use.

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

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

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

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

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

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

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

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

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.

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

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

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

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

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

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).

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

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

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

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

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

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

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

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

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

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.

'Antibodies, Neoplasm' is a medical term that refers to abnormal antibodies produced by neoplastic cells, which are cells that have undergone uncontrolled division and form a tumor or malignancy. These antibodies can be produced in large quantities and may have altered structures or functions compared to normal antibodies.

Neoplastic antibodies can arise from various types of malignancies, including leukemias, lymphomas, and multiple myeloma. In some cases, these abnormal antibodies can interfere with the normal functioning of the immune system and contribute to the progression of the disease.

In addition, neoplastic antibodies can also be used as tumor markers for diagnostic purposes. For example, certain types of monoclonal gammopathy, such as multiple myeloma, are characterized by the overproduction of a single type of immunoglobulin, which can be detected in the blood or urine and used to monitor the disease.

Overall, 'Antibodies, Neoplasm' is a term that encompasses a wide range of abnormal antibodies produced by neoplastic cells, which can have significant implications for both the diagnosis and treatment of malignancies.

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

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

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

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

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.

... (ADCC), also referred to as antibody-dependent cell-mediated cytotoxicity, is a ... "Antibody-dependent cell-mediated cytotoxicity against influenza virus-infected cells". The Journal of Infectious Diseases. 148 ... Wang, W; Erbe, AK; Hank, JA; Morris, ZS; Sondel, PM (2015). "NK Cell-Mediated Antibody-Dependent Cellular Cytotoxicity in ... ISBN 1-55581-246-5. University of Leicester, Virus Immunopathology Notes Antibody-Dependent+Cell+Cytotoxicity at the U.S. ...
Antibody-dependent cell-mediated cytotoxicity (ADCC) requires antibodies to bind to target cell surfaces. Antibodies are formed ... known as complement-dependent cytotoxicity; enhancement of antibody-dependent cell-mediated cytotoxicity; and CR3-dependent ... Fc receptors are found on many immune system cells, including NK cells. When NK cells encounter antibody-coated cells, the ... "Natural killer cell mediated antibody-dependent cellular cytotoxicity in tumor immunotherapy with therapeutic antibodies". ...
It triggers antibody-dependent cell-mediated cytotoxicity (ADCC). It does this by activating natural killer cells by binding to ... Drugs that are a monoclonal antibody, Monoclonal antibodies, Abandoned drugs, All stub articles, Monoclonal antibody stubs, ... CAIX is expressed on the surface of most renal cancer cells and is hypothesized to be on the surface of other tumor cells. It ... Girentuximab (Rencarex) for renal cell carcinoma v t e v t e (CS1 German-language sources (de), Drugs not assigned an ATC code ...
Lectin-8 Antibody that Induces Antibody-Dependent Cell-Mediated Cytotoxicity against Human Eosinophils and Inhibits Mast Cell- ... Lirentelimab depletes eosinophils via antibody-dependent natural killer cell mediated cytotoxicity. Lirentelimab is a humanized ... August 2020). "An anti-siglec-8 antibody depletes sputum eosinophils from asthmatic subjects and inhibits lung mast cells". ... Lirentelimab inhibits mast cells' IgE-mediated degranulation and de novo synthesis of prostaglandin D2 in vitro. Mild-to- ...
... or infected cells by antibody-mediated phagocytosis or antibody-dependent cell-mediated cytotoxicity. Some viruses such as ... This process is known as antibody-dependent cell-mediated cytotoxicity (ADCC). FcγRIII on NK cells can also associate with ... Another process involving Fc receptors is called antibody-dependent cell-mediated cytotoxicity (ADCC). During ADCC, FcγRIII ... CD4+ T cells (mature Th cells) provide help to B cells that produce antibodies. Several subsets of activated effector CD4+ T ...
This is thought to enhance its antibody-dependent cell-mediated cytotoxicity. It was first tested in humans in 2007. Kyowa ... Drugs that are a monoclonal antibody, Monoclonal antibodies, Orphan drugs, All stub articles, Monoclonal antibody stubs, ... Kyowa humanized it, and expressed the humanized gene in a CHO cell line in which FUT8 had been knocked out, which produced ... It was approved in Japan in 2012, for the treatment of relapsed or refractory CCR4+ adult T-cell leukemia/lymphoma (ATCLL) and ...
The contribution of antibody-dependent cell-mediated cytotoxicity to tumor cell killing can be measured with a specific test ... NK cells are thought to be an important cell type in this process. These cells are known as "uterine NK cells" (uNK cells) and ... CD56dim NK cells are always CD16 positive (CD16 is the key mediator of antibody-dependent cellular cytotoxicity (ADCC). ... Unmodified NK-92 cells lack CD-16, making them unable to perform antibody-dependent cellular cytotoxicity (ADCC); however, the ...
Meanwhile, antibody-dependent cell cytotoxicity (ADCC) remains unchanged in XLPDR NK cells. The most common manifestations of ... discovered that POLA1 deficiency is associated with decreased direct cytotoxicity of NK cells due to disturbances in vesicular ... and impaired direct cytotoxicity of NK cells are the most common symptoms. In females the disease is characterized by skin ... November 2019). "NK cell defects in X-linked pigmentary reticulate disorder". JCI Insight. 4 (21). doi:10.1172/jci.insight. ...
They cause thyroid cell damage by complement activation and antibody dependent cell cytotoxicity. However, anti-TPO antibodies ... The production of antibodies in Graves' disease is thought to arise by activation of CD4+ T-cells, followed by B-cell ... These B-cells produce antibodies specific to the thyroid antigens. In Hashimoto's thyroiditis, activated CD4+ T-cells produce ... of anti-TPO antibody positive cases also demonstrate thyroglobulin antibodies. Anti-Na+/I− symporter antibodies are a more ...
Antibody-dependent cell-mediated cytotoxicity: detection by automated flow cytometry with ultramicro techniques. Science. 1980 ... Prognostic and predictive value of a malignancy-risk gene signature in early-stage non-small cell lung cancer. Journal of the ... Increased Src activity disrupts cadherin/catenin-mediated homotypic adhesion in human colon cancer and transformed rodent cells ...
Helps kill breast cancer cells that overexpress HER-2, possibly through antibody-dependent cytotoxicity. Clinical use: ... Mechanism of action: A monoclonal antibody that binds to the glycoprotein receptor IIb/IIIa on activated platelets, preventing ... Predisposes to infections (reactivation of latent TB). Mechanism of action: A monoclonal antibody to CD20 surface ... Mechanism of action: A monoclonal antibody to TNF, proinflammatory cytokine. Clinical use: Crohn's disease, rheumatoid ...
... s are also capable of killing infected host cells via antibody-dependent cell-mediated cytotoxicity. Vacuolization may ... "Nr4a1-Dependent Ly6Clow Monocytes Monitor Endothelial Cells and Orchestrate Their Disposal". Cell. 153 (2): 362-375. doi: ... which activates CD4 Th2 cells and inhibits CD4 Th1 cell function. Many factors produced by other cells can regulate the ... With a diameter of 15-22 μm, monocytes are the largest cell type in peripheral blood. Monocytes are mononuclear cells and the ...
... complement-dependent cytotoxicity, antibody-dependent cell-mediated cytotoxicity and antibody-dependent phagocytosis. These ... Ansuvimab has been found to be capable of killing cells by antibody-dependent cell-mediated cytotoxicity. Other functional ... Antibodies are also able to "kill" virus particles directly and/or kill infected cells using antibody-mediated "effector ... The drug is composed of a single monoclonal antibody (mAb) and was initially isolated from immortalized B-cells that were ...
... cells. When expressed, CD32C plays an important role in the activation of antibody-dependent cell cytotoxicity (ADCC). Animal ... The therapeutic usage of monoclonal antibodies against CD32B can be effective for inducing cytotoxicity against B cell lymphoma ... Having too little CD32B has been associated with dysregulated antibody function, as well as increased antibody-dependent ... CD32 can be found on the surface of a variety of immune cells. CD32 has a low-affinity for the Fc region of IgG antibodies in ...
... by being deposited on the cell surface of the pathogen. In antibody-dependent cell-mediated cytotoxicity the pathogen does not ... This allows the antibody binding of an immune effector cell via its Fc domain. Antibody-dependent cell-mediated inherent ... The Fab portion of the antibody binds to the antigen, whereas the Fc portion of the antibody binds to an Fc receptor on the ... eosinophils and NK cells). Lack of mediation can cause inflammation of surrounding tissues and damage to healthy cells. Thau, L ...
Specifically, CD4 immunoadhesin plays a role in antibody-dependent cell-mediated cytotoxicity (ADCC) towards HIV-infected cells ... The Fc region of IgG1 is responsible for mediating effector functions such as antibody-dependent cell-mediated cytotoxicity ( ... While natural anti-gp120 antibodies exhibit a response towards uninfected CD4-expressing cells that have a soluble gp120 bound ... CD4-Ig binds to MHC class II molecules on antigen-presenting cells, thereby preventing the activation of T-helper cells that ...
October 2019). "Imlifidase Inhibits HLA Antibody-Mediated NK Cell Activation and Antibody-Dependent Cell-Mediated Cytotoxicity ... including CDC and antibody-dependent cell-mediated cytotoxicity (ADCC). Thus, imlifidase reduces the level of donor specific ... It cleaves the heavy chains of all human IgG subclasses (but no other immunoglobulins), eliminating Fc-dependent effector ... November 2018). "Safety, immunogenicity, pharmacokinetics, and efficacy of degradation of anti-HLA antibodies by IdeS ( ...
It is an unconjugated antibody, thought to work via the activation of antibody-dependent cell-mediated cytotoxicity (ADCC). It ... Drugs that are a monoclonal antibody, Monoclonal antibodies for tumors, Sanofi). ... It is a monoclonal antibody that binds to CD52, a protein present on the surface of mature lymphocytes, but not on the stem ... Hale G, Bright S, Chumbley G, Hoang T, Metcalf D, Munro AJ, Waldmann H (October 1983). "Removal of T cells from bone marrow for ...
This choice was made as TGN1112 showed antibody-dependent cellular cytotoxicity on CD28+ Jurkat cells. Thus the function of ... However, cell opsonisation by antibody leads normally to phagocytosis of the labeled cells, as seen in the case of HIV. In its ... Thus, attempts to induce FOXP3+ T cells might also induce effector cells capable of causing tissue damage. Other cells ... Since CD28 is the target of the TGN1412 antibody, M. fascicularis effector T-cells could not be stimulated by the drug. In 2013 ...
The mechanism of action includes B-cell lysis by antibody-dependent cellular cytotoxicity (ADCC), and complement-dependent ... complement-dependent cytotoxicity (CDC), and antibody-dependent cellular cytotoxicity (ADCC). Passive antibody therapy was ... complement-dependent cytotoxicity (CDC), and antibody-dependent cellular cytotoxicity (ADCC). Antagonism by antibodies ... For example, the ligation of CD40 monoclonal antibodies and CD40 on tumor cells license antigen-presenting cells (predominantly ...
... binds to BCMA on myeloma cell surfaces causing cell cycle arrest and inducing antibody-dependent cellular cytotoxicity. ... Belantamab mafodotin is a humanized IgG1κ monoclonal antibody against the B-cell maturation antigen (BCMA) conjugated with a ... Drugs that are a monoclonal antibody, Breakthrough therapy, GSK plc brands, Monoclonal antibodies for tumors, Orphan drugs, ... March 2019). "Antibody-drug conjugate, GSK2857916, in relapsed/refractory multiple myeloma: an update on safety and efficacy ...
... causing cells to apoptose via antibody-dependent cellular cytotoxicity, complement-dependent cytotoxicity, inhibition of ... Antibody-dependent cellular cytotoxicity is by means of natural killer cells. Unlike isatuximab which causes apoptosis directly ... Multiple myeloma cells with higher levels of CD38 show greater daratumumab-mediated cell lysis than cells with low CD38 ... which tends to mask the presence of any clinically significant antibodies. Treatment of the antibody panel cells with ...
... proliferation and survival of natural killer cells which carry out antibody-dependent cell-mediated cytotoxicity of tumor cells ... CD137 is expressed on the surfaces of activated CD8+ and CD4+ T cells, natural killer (NK) cells, dendritic cells, B cells, ... The receptor FcγRIIB is expressed on both CD8+ T cells and liver Kupffer cells. Because of this, CD8+ T cell activation by ... CD137 activation also promotes the proliferation and survival of B cells and CD4+ T cells, the differentiation of monocytes ...
... the antibody itself may trigger cell death via antibody-dependent cell-mediated cytotoxicity (ADCC), complement-dependent ... The antibody binds to the CD20 antigen found on the surface of normal and malignant B cells (but not B cell precursors), ... Drugs that are a monoclonal antibody, Monoclonal antibodies for tumors, Antibody-drug conjugates, Yttrium compounds, Indium ... Together, these actions eliminate B cells from the body, allowing a new population of healthy B cells to develop from lymphoid ...
The results of antibody-dependent cellular cytotoxicity assay on guineas-pig Kurloff cells strongly indicate an immunological ... Also, Kurloff cells present antibody-dependent cytotoxic activity in vitro. The structure of Kurloff cell was identified using ... "Antibody-dependent cellular cytotoxicity in the guinea pig: the role of the Kurloff cell". Cellular Immunology. 55 (2): 312-27 ... Antibody-Dependent Cellular Cytotoxicity (ADCC)", Antibody Fc, Academic Press, pp. 1-27, ISBN 978-0-12-394802-1, retrieved 2020 ...
... the efficacy of these antibodies in vivo is believed to be a result of antibody-dependent cell-mediated cytotoxicity and the ... "Vaccine-Induced Antibodies that Neutralize Group 1 and Group 2 Influenza A Viruses". Cell. 166 (3): 609-623. doi:10.1016/j.cell ... Therefore, HA is responsible for binding Influenza virus to sialic acid on the surface of target cells, such as cells in the ... Firstly, it allows the recognition of target vertebrate cells, accomplished through the binding to these cells' sialic acid- ...
These receptors bind to the Fc portion of IgG antibodies, which then activates antibody-dependent cell-mediated cytotoxicity ( ... involved in antibody-dependent cellular cytotoxicity (ADCC). It can be used to isolate populations of specific immune cells ... by immune complexes induces antibody-dependent cellular cytotoxicity (ADCC) in NK cells. However, this pathway can also be ... zeta complex in human natural killer cells. Induction by antibody-dependent cytotoxicity but not by natural killing". Journal ...
Antibody-dependent cell-mediated cytotoxicity (ADCC) describes the cell-killing ability of certain lymphocytes, which requires ... The live-cell protease is only active in cells that have a healthy cell membrane, and loses activity once the cell is ... Lymphocyte-mediated cytotoxicity, on the other hand, does not have to be mediated by antibodies; nor does complement-dependent ... Cytotoxicity is the quality of being toxic to cells. Examples of toxic agents are an immune cell or some types of venom, e.g. ...
Antibody dependent cell mediated cytotoxicity). It was shown that in order to initiate ADCC in vitro, PMN's have to adhere to ... On T cells and B cells, trogocytosis is triggered when the T cell receptor (TCR) on T cells or B cell receptor (BCR) on B cells ... from the cells they scan. The transfer was cell contact-dependent and occurred in the context of cell-conjugate formation. ... affinity T cell down-modulation of costimulatory molecules on dendritic cells mediated by T cells leads to regulation of T cell ...
... suggest a response of CR3 and CR4 to enable complement-dependent cell cytotoxicity towards antibody-coated cancer cells. Such ... they are involved in enhancing complement-dependent cytotoxicity in NK cells. Immunomodulatory therapies often aim for an ... phagocytosis and other cell-cell interactions in a variety of cells and circumstances. Upregulation of Mac-1 in the presence of ... Different faces of CR3 and CR4 in myeloid and lymphoid cells of mice and men" (PDF). Seminars in Cell & Developmental Biology. ...
... studies in our laboratory characterized a panel of highly mutated HIV-specific conformational epitope-targeting antibodies (Abs ... Antibody 2C6 Targets a Cross-Clade Conformational Epitope in gp41 with Highly Active Antibody-Dependent Cell Cytotoxicity J ... The significance of this site is supported by 2C6 having Ab-dependent cell cytotoxicity (ADCC) against envelope proteins from ... further definition and appreciation of targeting of antibodies similar to 2C6 during vaccine development should be considered. ...
NK cells directly identify and lyse cancer cells. Nascent transformed cells elicit NK cell activation and are eliminated. ... NK cells directly identify and lyse cancer cells. Nascent transformed cells elicit NK cell activation and are eliminated. ... but recent advances in elucidating NK cell biology have accelerated the development of NK cell-targeting therapeutics. NK cell ... but recent advances in elucidating NK cell biology have accelerated the development of NK cell-targeting therapeutics. NK cell ...
Novel TLR 7/8 agonists for improving NK cell mediated antibody-dependent cellular cytotoxicity (ADCC). In: Scientific reports. ... Novel TLR 7/8 agonists for improving NK cell mediated antibody-dependent cellular cytotoxicity (ADCC). Scientific reports. 2021 ... Novel TLR 7/8 agonists for improving NK cell mediated antibody-dependent cellular cytotoxicity (ADCC). / Khanna, Vidhi Devendra ... Novel TLR 7/8 agonists for improving NK cell mediated antibody-dependent cellular cytotoxicity (ADCC). ...
Antibody-dependent cell-mediated cytotoxicity triggered by dietary antigens in cows milk protein intolerance and coeliac ... such as immune complex-mediated complement activation or antibody-dependent cell-mediated cytotoxicity (ADCC). The biological ... in the presence of cytotoxic effector cells (NK cells or monocytes). Serum antibody levels of IgG, IgG subclasses and IgA ... Small amounts of dietary proteins pass undegraded into the circulation, an event initiating antibody production. Antibodies to ...
While glyco-engineered monoclonal antibodies (mAbs) with improved antibody-dependent cell-mediated cytotoxicity (ADCC). While ... glyco-engineered monoclonal antibodies (mAbs) with improved antibody-dependent cell-mediated cytotoxicity (ADCC) are achieving ... Cells were then infected with M13 helper phage (M13KO7, New Britain Biolabs, ratio bacterias: phage = 1:3) at 37 C for 20 min ... The human Fc gene was replicated with mutases using the 5 primer MG-619: 5-XL1-Blue cells and subsequently plated on solid 2YT ...
Antibody-dependent cell-mediated cytotoxicity (ADCC) requires antibodies to bind to target cell surfaces. Antibodies are formed ... known as complement-dependent cytotoxicity; enhancement of antibody-dependent cell-mediated cytotoxicity; and CR3-dependent ... Fc receptors are found on many immune system cells, including NK cells. When NK cells encounter antibody-coated cells, the ... "Natural killer cell mediated antibody-dependent cellular cytotoxicity in tumor immunotherapy with therapeutic antibodies". ...
Dive into the research topics of HIV-1 gp120-Specific Antibody-Dependent Cell-Mediated Cytotoxicity Correlates with Rate of ... HIV-1 gp120-Specific Antibody-Dependent Cell-Mediated Cytotoxicity Correlates with Rate of Disease Progression. ...
By blocking the cleavage of C5, this monoclonal antibody prevents cell damage caused by complement-mediated inflammation. We ... Antibodies, Monoclonal, Humanized / therapeutic use* * Antibody-Dependent Cell Cytotoxicity / drug effects * Atypical Hemolytic ... By blocking the cleavage of C5, this monoclonal antibody prevents cell damage caused by complement-mediated inflammation. We ... Loss of the therapeutic antibody via urine with concentrations up to 56 μg/ml correlated with proteinuria. In aHUS patients, ...
... demonstrated by antibody-dependent complement-mediated cytotoxicity.. Title. Cell surface display of the chlamydial glycolipid ... Antibody Specificity, Chlamydia, Complement System Proteins, Glycolipids, HeLa Cells, Humans, Polysaccharides, Bacterial. ... Thus, GLXA indeed is displayed on the surface of infected cells and, therefore, if antibody of appropriate specificity were ... pneumoniae-infected HeLa 229 cells, followed by the successive addition of mouse anti-GLXA antibody and complement, yielded ...
gamma delta T cell-meciated antibody-dependent cellular cytotoxicity with CD19 antibodies assessed by an impedance-based label- ... gamma delta T cell-meciated antibody-dependent cellular cytotoxicity with CD19 antibodies assessed by an impedance-based label- ... free real-time cytotoxicity assay. Author: Seidel, Ursula Joerdis Eva; Vogt, Fabian; Grosse-Hovest, Ludger; Jung, Gundram; ...
Antibody-dependent cell-mediated cytotoxicity (ADCC) assay. ADCC was determined by a calcein-acetyoxymethyl release assay ( ... Antibody-independent cell mediated cytotoxicity (AICC) was measured in wells containing target and effector cells without the ... but not patient serum exhibited hepatocyte killing efficacy in an antibody-dependent cell-mediated cytotoxicity (ADCC) assay. ... exhibited antibody-dependent cellular cytotoxicity of hepatocytes. Therefore, deposition of immunoglobulin antibodies may ...
Measurement of antibody-dependent cell-mediated cytotoxicity (ADCC). A role for ADCC in controlling initial development of HIV- ... Ab-dependent cell cytotoxicity in presence of NK cells was also investigated as well as aggregation and virus capture to assess ... including those dependent on interaction of Abs with Fc receptors such as antibody-dependent cellular cytotoxicity (ADCC), or ... Langerhans cells and interstitial or plasmacytoid dendritic cells. The function of anti-HIV antibodies was also dissected to ...
In addition, the inherited germline variants from these gene signatures were predominately enriched in T cell function, antigen ... and antibody-dependent cell-mediated cytotoxicity toward cancer cells.32 In breast cancer, MCs are linked with pro-angiogenic ... γδ T cells), resting natural killer cells (NK cells−), resting mast cells (MCs−), and CD8+ T cells (P = 3.14 × 10−2, P = 4.29 ... activated CD8 T cells (CD8+ T cells), T follicular helper cells (Tfh), monocytes (Monos), memory B cells, and activated B cells ...
Natural killer (NK) and antibody-dependent cell-mediated cytotoxicity (ADCC) activities as well as the number of large granular ... investigated whether VEGF affects the antibody-dependent cell-mediated cytotoxicity (ADCC) of human monocytes mediated by ... Natural Killer and Antibody-Dependent Cell-Mediated Cytotoxicity Activities and Large Granular Lymphocyte Frequencies in Viscum ... After short decreases during the first 24 h, significant increases in natural killer (NK) and antibody-dependent cell-mediated ...
... antibody-dependent cell cytotoxicity and phagocytosis, actually seem to favor cancer progression. In this review, we discuss ... antibody-dependent cell cytotoxicity and phagocytosis, actually seem to favor cancer progression. In this review, we discuss ... Despite the arsenal of defense strategies against foreign invaders, myeloid cells succumb to the instructions of an established ... Despite the arsenal of defense strategies against foreign invaders, myeloid cells succumb to the instructions of an established ...
Fluorescent antibody internalization assays work are key to determining the half-life of antibodies in the body based on the ... Carmen M et al: Antibody-dependent cell cytotoxicity: immunotherapy strategies enhancing effector NK cells. Immunology and Cell ... Figure 3. Antibody internalization response is cell number dependent. An increasing density of HT1080 cells were seeded (1-20 K ... T cell-like) and Raji (B cell-like) cells (30 K/well) were treated with different Incucyte® FabFluor labeled antibodies (4 μg/ ...
51Cr or more recently the 111In release assay was used to measure antibody dependent cell cytotoxicity. The target cells, ... Antibody Dependent Cell Cytotoxicity), immune complexes as well as circulating anti-TSA antibodies (mAb2 anti-idiotypes). A ... enhances antibody-dependent cellular cytotoxicity of human tumor cells mediated by human peripheral blood mononuclear cells. ... C. Method used to determine antibody dependent cell cytotoxicity (ADCC ). A 4 hr. ...
Gamma-delta T cells may be involved in antibody-dependent cell mediated cytotoxicity (ADCC). Compared with control subjects, ... Monoclonal antibodies to part of the 65-kd hsp of M tuberculosis react with Streptococcus sanguis. RAS thus may be a T cell- ... RAS probably involves cell-mediated mechanisms, but the precise immunopathogenesis remains unclear. Phagocytic and cytotoxic T ... Patients with active RAS have an increased proportion of gamma-delta T cells compared with control subjects and patients with ...
Antibody-dependent cell-mediated cytotoxicity. *Apoptosis. The exact contribution of each mechanism remains unclear, and ... How this occurs is unknown, but it involves T cells, B cells, and antibodies directed against the endothelial cells lining ... Plasma cells are B cells that do not usually express the CD20 antigen, so they mostly survive. As these produce antibodies, ... Cutaneous B cell lymphoma. Cutaneous B-cell lymphoma includes follicle centre, marginal zone, and diffuse large B-cell lymphoma ...
We first offer a general overview of DC biology and routes of Ag presentation eliciting effective T cell-mediated immune ... as a means to deliver antigen selectively to DCs and its effects on T-cell activation. We present an overview of the ... we review the fundamental aspects relevant for the development of cancer vaccines and the critical role of dendritic cells (DCs ... They found that Fc-μTP-L309C inhibited FcγR-mediated effector functions, such as antibody-dependent cell-mediated cytotoxicity ...
Through these mechanisms B cells are involved both in autoimmune diseases that are traditionally viewed as antibody mediated ... This paper includes an overview of the different functions of B cells in autoimmunity; the involvement of B cells in systemic ... This new understanding of the role of B cells opened up novel therapeutic options for the treatment of autoimmune diseases. ... We conclude with a discussion of novel therapies aimed at the selective targeting of pathogenic B cells. ...
Possible mechanisms of cell lysis include complement dependent cytotoxicity (CDC) and antibody dependent cell mediated ... showed signs of B-cell return with counts ,10 cells/μL. By Month 18, most patients (87%) had counts ,10 cells/μL. ... The detection of antibody formation is highly dependent on the sensitivity and specificity of the assay. Additionally, the ... A response to pneumococcal vaccination (a T-cell independent antigen) as measured by an increase in antibody titers to at least ...
... cells promoted transfer of the CAR cognate antigen from tumor to NK cells, resulting in (1) lower tumor antigen density, thus ... resulting in fratricide of trogocytic antigen-expressing NK cells (NKTROG+) and NK cell hyporesponsiveness. This phenomenon ... A new dual-chimeric antigen receptor (CAR) system enhances the antitumor activity of CAR natural killer cells and makes them ... signal to NK cells upon engagement with their TROG+ siblings. This system prevented trogocytic antigen-mediated fratricide, ...
Guidance on Antibody Dependent Cell-mediated Cytotoxicity (ADCC) activity. *Polysorbate, if applicable, in the finished product ...
CD4- and time-dependent susceptibility of HIV-1-infected cells to antibody-dependent cellular cytotoxicity. Lee, W. S., Prévost ... CXCR5+ follicular cytotoxic T cells control viral infection in B cell follicles. Leong, Y. A., Chen, Y., Ong, H. S., Wu, D., ... Low T-cell responses to mitogen stimulation predicts poor survival in recipients of allogeneic hematopoietic stem cell ... Anti-HIV antibody responses and the HIV reservoir size during antiretroviral therapy. Lee, S. A., Bacchetti, P., Chomont, N., ...
Bevacizumab is a humanized IgG1 monoclonal antibody that targets VEGF, a signaling protein that promotes the growth of new ... Bevacizumab displays no antibody-dependent cell-mediated cytotoxicity (ADCC) [4]. Bevacizumab has been approved by the FDA for ... This antibody has been removed from our catalog. However, you may contact us if you wish to order minimal quantities. ... Monoclonal human IgG4 (S228P) antibody against human VEGF. Anti-hVEGF-hIgG4 (S228P) features the constant region of the human ...
... the drug was shown to have very similar complement-dependent cytotoxicity, antibody-dependent cell-mediated cytotoxicity, and ... FDA Review Skeptical of ALS Stem Cell Therapy Ahead of Panel Meeting ...

No FAQ available that match "antibody dependent cell cytotoxicity"

No images available that match "antibody dependent cell cytotoxicity"