A member of the tumor necrosis factor receptor superfamily. It has specificity for LYMPHOTOXIN ALPHA1, BETA2 HETEROTRIMER and TUMOR NECROSIS FACTOR LIGAND SUPERFAMILY MEMBER 14. The receptor plays a role in regulating lymphoid ORGANOGENESIS and the differentiation of certain subsets of NATURAL KILLER T-CELLS. Signaling of the receptor occurs through its association with TNF RECEPTOR-ASSOCIATED FACTORS.
A membrane-bound tumor necrosis family member found primarily on LYMPHOCYTES. It can form a heterotrimer (LYMPHOTOXIN ALPHA1, BETA2 HETEROTRIMER) with the soluble ligand LYMPHOTOXIN-ALPHA and anchor it to the cell surface. The membrane-bound complex is specific for the LYMPHOTOXIN BETA receptor.
A tumor necrosis factor family member that is released by activated LYMPHOCYTES. Soluble lymphotoxin is specific for TUMOR NECROSIS FACTOR RECEPTOR TYPE I; TUMOR NECROSIS FACTOR RECEPTOR TYPE II; and TUMOR NECROSIS FACTOR RECEPTOR SUPERFAMILY, MEMBER 14. Lymphotoxin-alpha can form a membrane-bound heterodimer with LYMPHOTOXIN-BETA that has specificity for the LYMPHOTOXIN BETA RECEPTOR.
A member of tumor necrosis factor superfamily found on activated LYMPHOCYTES and MONOCYTES. It occurs as transmembrane protein that can be cleaved to release a secreted form that specifically binds to LYMPHOTOXIN BETA RECEPTOR and TUMOR NECROSIS FACTOR RECEPTOR SUPERFAMILY, MEMBER 14.
A heterotrimer complex consisting of one molecule of LYMPHOTOXIN-ALPHA and two molecules of the LYMPHOTOXIN-BETA. It is anchored to the cell surface via the transmembrane domains of the lymphotoxin-beta component and has specificity for the LYMPHOTOXIN BETA RECEPTOR. The lymphotoxin alpha1, beta2 heterotrimer plays a role in regulating lymphoid ORGANOGENESIS and the differentiation of certain subsets of NATURAL KILLER CELLS.
Cell surface receptors that bind TUMOR NECROSIS FACTORS and trigger changes which influence the behavior of cells.
A novel member of the tumor-necrosis factor receptor family that can also mediate HERPES SIMPLEX VIRUS TYPE 1 entry into cells. It has specificity for TUMOR NECROSIS FACTOR LIGAND SUPERFAMILY MEMBER 14 and the homotrimeric form of LYMPHOTOXIN-ALPHA. The receptor is abundantly expressed on T-LYMPHOCYTES and may play a role in regulating lymphocyte activation. Signaling by the activated receptor occurs through its association with TNF RECEPTOR-ASSOCIATED FACTORS.
A component of NF-kappa B transcription factor. It is proteolytically processed from NF-kappa B p100 precursor protein and is important for maturation of B-LYMPHOCYTES and adaptive HUMORAL IMMUNITY.
A CC-type chemokine with specificity for CCR7 RECEPTORS. It has activity towards DENDRITIC CELLS and T-LYMPHOCYTES.
Specialized tissues that are components of the lymphatic system. They provide fixed locations within the body where a variety of LYMPHOCYTES can form, mature and multiply. The lymphoid tissues are connected by a network of LYMPHATIC VESSELS.
Lymphoid tissue on the mucosa of the small intestine.
A tumor necrosis factor receptor subtype that has specificity for TUMOR NECROSIS FACTOR ALPHA and LYMPHOTOXIN ALPHA. It is constitutively expressed in most tissues and is a key mediator of tumor necrosis factor signaling in the vast majority of cells. The activated receptor signals via a conserved death domain that associates with specific TNF RECEPTOR-ASSOCIATED FACTORS in the CYTOPLASM.
Strains of mice in which certain GENES of their GENOMES have been disrupted, or "knocked-out". To produce knockouts, using RECOMBINANT DNA technology, the normal DNA sequence of the gene being studied is altered to prevent synthesis of a normal gene product. Cloned cells in which this DNA alteration is successful are then injected into mouse EMBRYOS to produce chimeric mice. The chimeric mice are then bred to yield a strain in which all the cells of the mouse contain the disrupted gene. Knockout mice are used as EXPERIMENTAL ANIMAL MODELS for diseases (DISEASE MODELS, ANIMAL) and to clarify the functions of the genes.
Serum glycoprotein produced by activated MACROPHAGES and other mammalian MONONUCLEAR LEUKOCYTES. It has necrotizing activity against tumor cell lines and increases ability to reject tumor transplants. Also known as TNF-alpha, it is only 30% homologous to TNF-beta (LYMPHOTOXIN), but they share TNF RECEPTORS.
Inbred C57BL mice are a strain of laboratory mice that have been produced by many generations of brother-sister matings, resulting in a high degree of genetic uniformity and homozygosity, making them widely used for biomedical research, including studies on genetics, immunology, cancer, and neuroscience.
The 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.
Proteins which are found in membranes including cellular and intracellular membranes. They consist of two types, peripheral and integral proteins. They include most membrane-associated enzymes, antigenic proteins, transport proteins, and drug, hormone, and lectin receptors.
Cell-surface proteins that bind transforming growth factor beta and trigger changes influencing the behavior of cells. Two types of transforming growth factor receptors have been recognized. They differ in affinity for different members of the transforming growth factor beta family and in cellular mechanisms of action.
Non-hematopoietic cells, with extensive dendritic processes, found in the primary and secondary follicles of lymphoid tissue (the B cell zones). They are different from conventional DENDRITIC CELLS associated with T-CELLS. They are derived from MESENCHYMAL STEM CELLS and are negative for class II MHC antigen and do not process or present antigen like the conventional dendritic cells do. Instead, follicular dendritic cells have FC RECEPTORS and C3B RECEPTORS that hold antigen in the form of ANTIGEN-ANTIBODY COMPLEXES on their surfaces for long periods for recognition by B-CELLS.
A PDGF receptor that binds specifically to the PDGF-B chain. It contains a protein-tyrosine kinase activity that is involved in SIGNAL TRANSDUCTION.
A transcription factor that takes part in the NF-kappa-B complex by interacting with NF-KAPPA B P50 SUBUNIT or NF-KAPPA B P52 SUBUNIT. It regulates GENETIC TRANSCRIPTION that is involved in immune and inflammatory responses.

Signalling by CD95 and TNF receptors: not only life and death. (1/218)

Members of the TNF family of receptors play important roles in normal physiology and in defence. The recent rapid progress in the understanding of the mechanisms of apoptosis has been accompanied by assumptions that TNF family receptors such as CD95(Fas/APO-1) only have a role in regulating cell survival. While regulation of cell death is one important function of TNF family receptors, they are capable of activating signal transduction pathways that have many other effects. The present review will focus on signalling of some TNF family receptors in the immune system, not only for apoptosis, but also for survival or activation.  (+info)

Expression of the lymphotoxin beta receptor on follicular stromal cells in human lymphoid tissues. (2/218)

The lymphotoxin beta receptor (LTbetaR), and its ligand, LTalpha1beta2, have been proposed to play a key role in the development and organization of lymphoid tissues. The LTbetaR is expressed on a variety of human primary and transformed cells, but strikingly absent on T or B lymphocytes and primary monocytes or peripheral dendritic cells, although LTbetaR is detected on some myeloid leukemic lines. In the developing thymus LTbetaR is prominent along the trabeculae and into the medulla upto corticomedullary junction. In the spleen, LTbetaR is prominently expressed by cells in the red pulp and along the borders of red and white pulp which colocalizes with reticular stromal cells. The LTbetaR is expressed on a human follicular dendritic cell line, FDC-1, and signals expression of CD54 when ligated with the LTalpha1beta2 complex. These results support the concept that directional interactions between LTalpha1beta2 bearing lymphocytes and LTbetaR bearing stromal cells are involved in the organization of lymphoid tissue.  (+info)

The cytoplasmic domain of the lymphotoxin-beta receptor mediates cell death in HeLa cells. (3/218)

Activation of lymphotoxin-beta receptor (LT-betaR) by conjugation with heterotrimeric lymphotoxin, LT-alpha1/beta2, or by cross-linking with anti-LT-betaR antibodies can trigger apoptosis. We have observed that overexpression of either LT-betaR or the cytoplasmic domain of LT-betaR (LT-betaR(CD)) also induces apoptosis, which may be attributed to the tendency of LT-betaR(CD) to self-associate. The self-association domain of LT-betaR(CD) was mapped to amino acids 324-377, a region of the protein that is also essential for LT-betaR-induced apoptosis. Furthermore, we have shown that LT-betaR(CD)-induced apoptosis could be inhibited by a TRAF3 dominant negative mutant and by the caspase inhibitors Z-VAD-FMK, DEVD-FMK, and CrmA. The ligand-independent apoptosis induced by LT-betaR(CD) will help us to further dissect LT-betaR signaling pathway.  (+info)

Signaling through the lymphotoxin-beta receptor stimulates HIV-1 replication alone and in cooperation with soluble or membrane-bound TNF-alpha. (4/218)

The level of ongoing HIV-1 replication within an individual is critical to HIV-1 pathogenesis. Among host immune factors, the cytokine TNF-alpha has previously been shown to increase HIV-1 replication in various monocyte and T cell model systems. Here, we demonstrate that signaling through the TNF receptor family member, the lymphotoxin-beta (LT-beta) receptor (LT-betaR), also regulates HIV-1 replication. Furthermore, HIV-1 replication is cooperatively stimulated when the distinct LT-betaR and TNF receptor systems are simultaneously engaged by their specific ligands. Moreover, in a physiological coculture cellular assay system, we show that membrane-bound TNF-alpha and LT-alpha1beta2 act virtually identically to their soluble forms in the regulation of HIV-1 replication. Thus, cosignaling via the LT-beta and TNF-alpha receptors is probably involved in the modulation of HIV-1 replication and the subsequent determination of HIV-1 viral burden in monocytes. Intriguingly, surface expression of LT-alpha1beta2 is up-regulated on a T cell line acutely infected with HIV-1, suggesting a positive feedback loop between HIV-1 infection, LT-alpha1beta2 expression, and HIV-1 replication. Given the critical role that LT-alpha1beta2 plays in lymphoid architecture, we speculate that LT-alpha1beta2 may be involved in HIV-associated abnormalities of the lymphoid organs.  (+info)

Involvement of distinct cellular compartments in the abnormal lymphoid organogenesis in lymphotoxin-alpha-deficient mice and alymphoplasia (aly) mice defined by the chimeric analysis. (5/218)

Both lymphotoxin-alpha (LTalpha)-deficient mice and alymphoplasia (aly) mice, a natural mutant strain, manifest a quite similar phenotype: lack of lymph nodes (LN) and Peyer's patches (PP), with disturbed spleen architecture. The mechanisms underlying the defective lymphoid organogenesis in these mice were investigated by generating aggregation chimeras; ex vivo fused morulae were implanted into pseudo-pregnant host females and allowed to develop to term. Chimeric mice between LTalpha-deficient mice and wild-type mice restored LN and PP almost completely, suggesting that LTalpha expressed by circulating bone marrow-derived cells is essential for lymphoid organogenesis as well as for organization of spleen architecture. By contrast, chimeric mice between aly mice and wild-type mice showed only limited restoration of LN and PP. This suggests that the putative aly gene product does not act as a circulating ligand for lymphoid organogenesis, like LTalpha. Rather, abnormal development of lymphoid organs in aly mice seems most likely due to the defective development of the incipient stromal cells of the LN and PP. Supporting this hypothesis, up-regulation of VCAM-1 on aly mouse embryonic fibroblasts by signals through LTbetaR, which is exclusively expressed by nonlymphoid cells, was disturbed. These studies demonstrate that LTalpha and the putative aly gene product together control lymphoid organogenesis with a close mechanistic relationship in their biochemical pathways through governing the distinct cellular compartments, the former acting as a circulating ligand and the latter as a LTbetaR-signaling molecule expressed by the stroma of the lymphoid organs.  (+info)

The requirement of membrane lymphotoxin for the presence of dendritic cells in lymphoid tissues. (6/218)

Although several cytokines, including tumor necrosis factor (TNF), can promote the growth of dendritic cells (DCs) in vitro, the cytokines that naturally regulate DC development and function in vivo have not been well defined. Here, we report that membrane lymphotoxin (LT), instead of TNF, regulates the migration of DCs in the spleen. LTalpha(-/-) mice, lacking membrane LTalpha/beta and LTalpha(3), show markedly reduced numbers of DCs in the spleen. Unlike wild-type mice and TNF(-/-) mice that have densely clustered DCs in the T cell zone and around the marginal zone, splenic DCs in LTalpha(-/-) mice are randomly distributed. The reduced number of DCs in lymphoid tissues of LTalpha(-/-) mice is associated with an increased number of DCs in nonlymphoid tissues. The number of splenic DCs in LTalpha(-/-) mice is restored when additional LT-expressing cells are provided. Blocking membrane LTalpha/beta in wild-type mice markedly diminishes the accumulation of DCs in lymphoid tissues. These data suggest that membrane LT is an essential ligand for the presence of DCs in the spleen. Mice deficient in TNF receptor, which is the receptor for both soluble LTalpha(3) and TNF-alpha(3) trimers, have normal numbers of DCs. However, LTbetaR(-/-) mice show reduced numbers of DCs, similar to the mice lacking membrane LT alpha/beta. Taken together, these results support the notion that the signaling via LTbetaR by membrane LTalpha/beta is required for the presence of DCs in lymphoid tissues.  (+info)

Up-regulation of BOB.1/OBF.1 expression in normal germinal center B cells and germinal center-derived lymphomas. (7/218)

The BOB.1/OBF.1/OCAB.1 protein is a lymphocyte-specific transcriptional coactivator. It interacts with the Oct1 and Oct2 transcription factors and contributes to the transcriptional activity of octamer motifs. The analysis of established B cell lines had suggested that BOB.1/OBF.1 is constitutively expressed at all stages of B cell development. Here we show that expression of BOB. 1/OBF.1 is regulated within the B cell lineage. Specifically, germinal center B cells show highly increased BOB.1/OBF.1 levels. We can induce the up-regulation by stimulating primary splenic B cells, eg, by triggering CD40 signaling in the presence of interleukin-4. Expression of BOB.1/OBF.1 is detectable but reduced in spleens from mice unable to undergo the germinal center reaction due to mutations in the TNF receptor p55 or lymphotoxin beta (LTbeta) receptor genes. Furthermore, we demonstrate that BOB.1/OBF.1 expression is highly regulated in human B cell lymphomas. Whereas lymphomas representing pre- and postfollicular B cell developmental stages are negative for BOB.1/OBF.1, high-level expression of BOB.1/OBF.1 is characteristic of germinal center-derived tumors. In these tumors BOB.1/OBF.1 is typically coexpressed with high levels of Bcl6. These results imply that overexpression of BOB.1/OBF.1, like overexpression of Bcl6, might play a role in the pathogenesis of germinal center-derived B cell lymphomas. Furthermore, overexpression of BOB.1/OBF.1 represents a characteristic feature of these tumors that is useful in their identification.  (+info)

Discrete signaling regions in the lymphotoxin-beta receptor for tumor necrosis factor receptor-associated factor binding, subcellular localization, and activation of cell death and NF-kappaB pathways. (8/218)

Lymphotoxin-beta receptor (LTbetaR), a member of the tumor necrosis factor receptor superfamily, is essential for the development and organization of secondary lymphoid tissue. Wild type and mutant LTbetaR containing successive truncations of the cytoplasmic domain were investigated by retrovirus-mediated gene transfer into HT29.14s and in 293T cells by transfection. Wild type receptors accumulated in perinuclear compartments and enhanced responsiveness to ligand-induced cell death and ligand-independent activation of NFkappaB p50 dimers. Coimmunoprecipitation and confocal microscopy mapped the TRAF3 binding site to amino acids PEEGDPG at position 389. However, LTbetaR truncated at position Pro(379) acted as a dominant positive mutant that down-modulated surface expression and recruited TRAF3 to endogenous LTbetaR. This mutant exhibited ligand-independent cell death and activated NF-kappaB p50 dimers. By contrast, truncation at Gly(359) created a dominant-negative mutant that inhibited ligand-induced cell death and activation of NF-kappaB p50/p65 heterodimers. This mutant also blocked accumulation of wild type receptor into perinuclear compartments, suggesting subcellular localization may be crucial for signal transduction. A cryptic TRAF-independent NF-kappaB activating region was identified. These mutants define discrete subregions of a novel proline-rich domain that is required for subcellular localization and signal transduction by the LTbetaR.  (+info)

The Lymphotoxin-beta receptor (LTβR) is a type III transmembrane protein and a member of the tumor necrosis factor receptor superfamily (TNFRSF). It is primarily expressed on the surface of various cell types, including immune cells such as lymphocytes, dendritic cells, and stromal cells in lymphoid organs.

LTβR binds to its ligands, Lymphotoxin-alpha (LTα) and Lymphotoxin-beta (LTβ), which are primarily produced by activated T-cells and B-cells. The binding of LTα/LTβ to LTβR triggers a signaling cascade that leads to the activation of various downstream signaling pathways, including NF-κB and MAPK pathways.

The activation of LTβR plays critical roles in the development and organization of lymphoid tissues, immune responses, and inflammation. Dysregulation of LTβR signaling has been implicated in various autoimmune diseases, such as rheumatoid arthritis, inflammatory bowel disease, and multiple sclerosis.

Lymphotoxin-beta (LT-β) is a cytokine that belongs to the tumor necrosis factor (TNF) family. It is primarily produced by activated T lymphocytes and plays an essential role in the development and organization of the immune system, particularly in the formation of lymphoid structures such as lymph nodes and Peyer's patches.

LT-β forms a complex with its receptor, LTβR, which is expressed on various cell types including stromal cells, endothelial cells, and immune cells. The binding of LT-β to LTβR triggers a cascade of intracellular signaling events that lead to the activation of genes involved in the development and maintenance of lymphoid tissues.

In addition to its role in lymphoid organogenesis, LT-β has been implicated in various immune responses, including inflammation, immune cell recruitment, and the regulation of adaptive immunity. Dysregulation of LT-β signaling has been associated with several autoimmune diseases, making it a potential target for therapeutic intervention.

Lymphotoxin-alpha (LT-alpha), also known as Tumor Necrosis Factor-beta (TNF-beta), is a cytokine that belongs to the TNF superfamily. It is primarily produced by activated CD4+ and CD8+ T cells, and to some extent by B cells, natural killer (NK) cells, and neutrophils. LT-alpha can form homotrimers or heterotrimers with Lymphotoxin-beta (LT-beta), which bind to the LT-beta receptor (LTβR) and herceptin-resistant tumor cells (HRT) on the surface of various cell types, including immune cells, fibroblasts, and endothelial cells.

The activation of the LTβR signaling pathway plays a crucial role in the development and organization of secondary lymphoid organs, such as lymph nodes, Peyer's patches, and spleen. Additionally, LT-alpha has proinflammatory effects, inducing apoptosis in susceptible cells, activating immune cells, and contributing to the pathogenesis of several inflammatory diseases, including rheumatoid arthritis, psoriasis, and Crohn's disease.

Tumor Necrosis Factor Ligand Superfamily Member 14 (TNFSF14), also known as HVEM (Herpesvirus Entry Mediator) Ligand or Lymphotoxin-like, Inhibitory or Secreting Factor (LIGHT), is a type II transmembrane protein and a member of the Tumor Necrosis Factor (TNF) ligand superfamily. It plays a crucial role in immune cell communication and regulation of inflammatory responses.

TNFSF14 can exist as both a membrane-bound form and a soluble form, produced through proteolytic cleavage or alternative splicing. The protein interacts with two receptors: HVEM (TNFRSF14) and Lymphotoxin β Receptor (LTβR). Depending on the receptor it binds to, TNFSF14 can have either costimulatory or inhibitory effects on immune cell functions.

The binding of TNFSF14 to HVEM promotes the activation and proliferation of T cells, enhances the cytotoxic activity of natural killer (NK) cells, and contributes to the development and maintenance of secondary lymphoid organs. In contrast, the interaction between TNFSF14 and LTβR primarily induces the formation and remodeling of tertiary lymphoid structures in peripheral tissues during inflammation or infection.

Dysregulation of TNFSF14 has been implicated in various pathological conditions, including autoimmune diseases, chronic inflammation, and cancer. Therefore, targeting this molecule and its signaling pathways is an area of interest for developing novel therapeutic strategies to treat these disorders.

Lymphotoxin-alpha1, beta2 heterotrimer (LT-alpha1/beta2) is not typically defined in the context of medical terminology. However, it is a term used to describe a specific form of lymphotoxin, which is a cytokine involved in the immune response.

Lymphotoxins are a type of tumor necrosis factor (TNF) that can induce apoptosis (programmed cell death) in certain cells. They exist in two forms: LT-alpha and LT-beta, which can combine to form heterotrimers (LT-alpha1/beta2) or homotrimers (LT-alpha3 or LT-beta3).

The LT-alpha1/beta2 heterotrimer is a transmembrane protein complex composed of one alpha and two beta subunits. It plays an important role in the development and maintenance of lymphoid tissue, including the formation of germinal centers in lymph nodes and Peyer's patches.

In summary, while not a strictly medical definition, Lymphotoxin-alpha1, beta2 heterotrimer refers to a specific form of lymphotoxin that plays a crucial role in the immune response by contributing to the development and maintenance of lymphoid tissue.

Tumor Necrosis Factor (TNF) Receptors are cell surface receptors that bind to tumor necrosis factor cytokines. They play crucial roles in the regulation of a variety of immune cell functions, including inflammation, immunity, and cell survival or death (apoptosis).

There are two major types of TNF receptors: TNFR1 (also known as p55 or CD120a) and TNFR2 (also known as p75 or CD120b). TNFR1 is widely expressed in most tissues, while TNFR2 has a more restricted expression pattern and is mainly found on immune cells.

TNF receptors have an intracellular domain called the death domain, which can trigger signaling pathways leading to apoptosis when activated by TNF ligands. However, they can also activate other signaling pathways that promote cell survival, differentiation, and inflammation. Dysregulation of TNF receptor signaling has been implicated in various diseases, including cancer, autoimmune disorders, and neurodegenerative conditions.

Tumor necrosis factor receptor superfamily member 14 (TNFRSF14), also known as HVEM (herpesvirus entry mediator), is a type of cell surface receptor that belongs to the tumor necrosis factor receptor superfamily. It is involved in various immune responses and can be found on the surface of different types of cells, including T cells, B cells, and myeloid cells.

TNFRSF14 has been shown to interact with several ligands, including LIGHT (TNFSF14) and BTLA (B- and T-lymphocyte attenuator), which can either activate or inhibit immune responses. The interaction between TNFRSF14 and its ligands plays a crucial role in regulating the activation, proliferation, and effector functions of immune cells.

In the context of tumors, TNFRSF14 has been found to be expressed on some tumor cells, where it can contribute to tumor growth and progression by promoting immune evasion and resistance to therapies. Additionally, genetic variations in TNFRSF14 have been associated with susceptibility to certain autoimmune diseases, such as rheumatoid arthritis and systemic lupus erythematosus.

Overall, TNFRSF14 is a critical regulator of immune responses and has important implications for the development of cancer and autoimmune diseases.

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

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

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

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

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

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

Lymphoid tissue is a specialized type of connective tissue that is involved in the immune function of the body. It is composed of lymphocytes (a type of white blood cell), which are responsible for producing antibodies and destroying infected or cancerous cells. Lymphoid tissue can be found throughout the body, but it is particularly concentrated in certain areas such as the lymph nodes, spleen, tonsils, and Peyer's patches in the small intestine.

Lymphoid tissue provides a site for the activation, proliferation, and differentiation of lymphocytes, which are critical components of the adaptive immune response. It also serves as a filter for foreign particles, such as bacteria and viruses, that may enter the body through various routes. The lymphatic system, which includes lymphoid tissue, helps to maintain the health and integrity of the body by protecting it from infection and disease.

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

Tumor Necrosis Factor Receptor 1 (TNFR1), also known as p55 or CD120a, is a type I transmembrane protein that belongs to the tumor necrosis factor receptor superfamily. It is widely expressed in various tissues and cells, including immune cells, endothelial cells, and fibroblasts. TNFR1 plays a crucial role in regulating inflammation, immunity, cell survival, differentiation, and apoptosis (programmed cell death).

TNFR1 is activated by its ligand, Tumor Necrosis Factor-alpha (TNF-α), which is a potent proinflammatory cytokine produced mainly by activated macrophages and monocytes. Upon binding of TNF-α to TNFR1, a series of intracellular signaling events are initiated through the recruitment of adaptor proteins, such as TNF receptor-associated death domain (TRADD), receptor-interacting protein kinase 1 (RIPK1), and TNF receptor-associated factor 2 (TRAF2). These interactions lead to the activation of several downstream signaling pathways, including nuclear factor kappa B (NF-κB) and mitogen-activated protein kinases (MAPKs), which ultimately regulate gene expression and cellular responses.

TNFR1 has been implicated in various physiological and pathological processes, such as inflammation, infection, autoimmunity, cancer, and neurodegenerative disorders. Dysregulation of TNFR1 signaling can contribute to the development and progression of several diseases, making it an attractive target for therapeutic interventions.

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

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

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

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

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

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.

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.

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

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

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

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

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

Transforming Growth Factor beta (TGF-β) receptors are a group of cell surface receptors that bind to TGF-β ligands and transduce signals into the cell. These receptors play crucial roles in regulating various cellular processes, including cell growth, differentiation, apoptosis, and extracellular matrix production.

There are two types of TGF-β receptors: type I and type II. Type I receptors, also known as activin receptor-like kinases (ALKs), have serine/threonine kinase activity and include ALK1, ALK2, ALK3, ALK4, ALK5, and ALK6. Type II receptors are constitutively active serine/threonine kinases and include TGF-β RII, ActRII, and ActRIIB.

When a TGF-β ligand binds to a type II receptor, it recruits and phosphorylates a type I receptor, which in turn phosphorylates downstream signaling molecules called Smads. Phosphorylated Smads form complexes with co-Smad proteins and translocate to the nucleus, where they regulate gene expression.

Abnormalities in TGF-β signaling have been implicated in various human diseases, including fibrosis, cancer, and autoimmune disorders. Therefore, understanding the mechanisms of TGF-β receptor function is essential for developing therapeutic strategies to target these conditions.

Follicular dendritic cells (FDCs) are a specialized type of dendritic cell that reside in the germinal centers of secondary lymphoid organs, such as the spleen, lymph nodes, and Peyer's patches. They play a critical role in the adaptive immune response by presenting antigens to B cells and helping to regulate their activation, differentiation, and survival.

FDCs are characterized by their extensive network of dendrites, which can trap and retain antigens on their surface for extended periods. They also express a variety of surface receptors that allow them to interact with other immune cells, including complement receptors, Fc receptors, and cytokine receptors.

FDCs are derived from mesenchymal stem cells and are distinct from classical dendritic cells, which are derived from hematopoietic stem cells. They are long-lived cells that can survive for months or even years in the body, making them important players in the maintenance of immune memory.

Overall, follicular dendritic cells play a critical role in the adaptive immune response by helping to regulate B cell activation and differentiation, and by contributing to the development of immune memory.

The platelet-derived growth factor beta (PDGF-β) receptor is a type of cell surface receptor that binds to specific proteins called platelet-derived growth factors (PDGFs). PDGFs are important signaling molecules involved in various biological processes, including cell growth, division, and survival.

The PDGF-β receptor is a transmembrane protein with an extracellular domain that binds to PDGFs and an intracellular domain that activates downstream signaling pathways when activated by PDGF binding. The PDGF-BB isoform specifically binds to the PDGF-β receptor, leading to its activation and initiation of signaling cascades that promote cell proliferation, migration, and survival.

Mutations in the PDGF-β receptor gene have been associated with certain types of cancer and vascular diseases, highlighting its importance in regulating cell growth and division. Inhibitors of the PDGF-β receptor have been developed as potential therapeutic agents for the treatment of various cancers and other diseases.

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

  • Lymphotoxin beta receptor (LTBR), also known as tumor necrosis factor receptor superfamily member 3 (TNFRSF3), is a cell surface receptor for lymphotoxin involved in apoptosis and cytokine release. (wikipedia.org)
  • One particular modifier gene, lymphotoxin beta receptor (LTBR), acts like a molecular fountain of youth: with LTBR, T cells multiply, have a greater proportion of younger, more stem cell-like cells and resist becoming exhausted over time. (globalhealthnewswire.com)
  • It is a member of the tumor necrosis factor receptor superfamily. (wikipedia.org)
  • Human Lymphotoxin beta receptor (L Tbeta R, TNFRSF3) is a member of the TNF receptor superfamily with similarilty to CD120a(TNFR1) and CD120b(TNFR2). (ancell.com)
  • The lymphotoxin beta receptor (LTβR) belongs to the TNF receptor superfamily and is essential for the organogenesis of secondary lymphoid organs as well as the coordination of an effective immune response against invading pathogens. (sfi-dgfi-2023.fr)
  • The monoclonal antibody 55R-170 recognizes the extracellular part of mouse Tumor Necrosis Factor Receptor superfamily member 1A (TNF-RI), also known as CD120a or p55. (hycultbiotech.com)
  • Ligands for these receptors belong to the Tumor Necrosis Factor (TNF) superfamily of cytokines, which activate signaling pathways for cell survival, death, and differentiation that orchestrate the development, organization and homeostasis of lymphoid, mammary, neuronal and ectodermal tissues. (hycultbiotech.com)
  • A member of the tumor necrosis factor receptor superfamily found on most T-LYMPHOCYTES. (bvsalud.org)
  • In various proinflammatory cytokine-stimulated cells, ligand-receptor interactions initially activate TAK1. (xiahepublishing.com)
  • Lymphotoxin alpha, a member of the tumor necrosis factor family , is a cytokine produced by lymphocytes . (wikidoc.org)
  • Infliximab neutralizes cytokine TNF-alpha and inhibits its binding to TNF-alpha receptor. (medscape.com)
  • LIGHT binds to two different receptors, Herpes Virus Entry Mediator (HVEM) and Lymphotoxin beta Receptor (LTβR). (amjournals.org)
  • TNF-RI belongs to the large TNF receptor family, among which TNF-RII (TNF-R p75-80), lymphotoxin-beta receptor (LTbetaR) and the Herpes virus entry mediator (HVEM). (hycultbiotech.com)
  • Here we report that when mice lacking the RelA subunit of NF-κB are brought to term by breeding onto a tumor necrosis factor receptor (TNFR)1-deficient background, the mice that are born lack lymph nodes, Peyer's patches, and an organized splenic microarchitecture, and have a profound defect in T cell-dependent antigen responses. (rupress.org)
  • Secreted autoantibodies specific to receptors or receptor ligands can activate or inhibit receptor functions. (hindawi.com)
  • In activating cells, NF-κB signaling is activated through a series of signaling cascades, following the ligation of various cell surface receptors with paired ligands. (xiahepublishing.com)
  • 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. (lookformedical.com)
  • 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. (lookformedical.com)
  • Deletion of the death domain of the TNF-RI results in a non-functional receptor, indicating that the death domain is required for the signal transduction of the physiological functions of TNF-RI in vivo. (hycultbiotech.com)
  • Recruitment and activation of naive T cells in the islets by lymphotoxin beta receptor-dependent tertiary lymphoid structure. (uchicago.edu)
  • He has completed his university studies with a Master degree in "Medical Biotechnology" still at Sapienza university of Rome with a thesis entitled: Role of hMENA isoforms on tertiary lymphoid structures localization and lymphotoxin beta receptor regulation in lung carcinoma (2018/2019). (capstone-etn.eu)
  • Using an adenoviral vector encoding kinase-deficient NIK, we have investigated the role of NIK in LPS, IL-1, TNF-alpha, and lymphotoxin (LT) betaR signaling in primary human cells and TNF-alpha expression from rheumatoid tissue. (ox.ac.uk)
  • Suppression of LT beta R signaling can alleviate autoimmunity(2) or exasperate mycobacterial infection(6). (ancell.com)
  • By employing single-cell transcriptomics, endothelial fate mapping, and functional multiplex immune profiling, we demonstrate that antiangiogenic immune-modulating therapies evoke transdifferentiation of postcapillary venules into inflamed high-endothelial venules (HEVs) via lymphotoxin/lymphotoxin beta receptor (LT/LTβR) signaling. (utu.fi)
  • Fig. 1 Canonical and noncanonical NF-κB signaling pathways in resting vs. receptor-stimulated cells. (xiahepublishing.com)
  • Formation of LT-α 1 -β 2 complex enables binding to LT-β receptors and subsequent activation of signaling pathways. (wikidoc.org)
  • Here, we show that a group of neurons in the Drosophila larval brain expresses the adiponectin receptor (AdipoR) and controls systemic growth and metabolism through insulin signaling. (cnrs.fr)
  • Signaling by the activated receptor occurs through its association with TNF RECEPTOR-ASSOCIATED FACTORS. (bvsalud.org)
  • Interferons exert their pleiotropic effects through two different receptors. (lookformedical.com)
  • CAR-T cells have antigen receptors on their surface that recognize specific proteins present on cancer cells to target and destroy them. (globalhealthnewswire.com)
  • The new method that they developed - OverCITE-seq - allowed the researchers to test the impact of different modifier genes on the cellular states of T cells, which includes the expression of every gene, the proteins decorating the cell surface, and the unique T cell receptors expressed by each cell (clonotype). (globalhealthnewswire.com)
  • simultaneously bind to major histocompatibility complex published a study of seven patients with severe streptococ- class II molecules and T-cell receptor molecules bearing a cal infections: SPE-A was detected in serum samples from four patients (27). (cdc.gov)
  • Here we demonstrate that in mice treated with FTY720 or that lack sphingosine-1-phosphate (S1P) receptor-1 (S1P 1 ) in B cells, IgG ASCs are induced and localize normally in secondary lymphoid organs but they are reduced in numbers in blood and BM. (silverchair.com)
  • Since first developed more than 30 years ago, chimeric antigen receptor (CAR)-T cell therapy has proven highly effective in targeting blood cancer cells, resulting in multiple FDA-approved CAR-T therapies. (globalhealthnewswire.com)
  • The protein specifically binds the lymphotoxin membrane form (a complex of lymphotoxin-alpha and lymphotoxin-beta). (wikipedia.org)
  • As a member of the TNF family , LT-α binds to various receptors and activates the NF-κB pathway , thus promoting immune regulation through the innate immune response. (wikidoc.org)
  • An interferon regulatory factor that binds upstream TRANSCRIPTIONAL REGULATORY ELEMENTS in the GENES for INTERFERON-ALPHA and INTERFERON-BETA . (lookformedical.com)
  • 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. (lookformedical.com)
  • The protein encoded by this gene is a member of the tumor necrosis factor (TNF) family of receptors. (wikipedia.org)
  • A specific S. salivarius protein, RSSL-01370, contains regions with homology to the Spike receptor-binding domain, and immunization of mice with RSSL-01370 elicited anti-Spike IgG antibodies in the serum. (bvsalud.org)
  • Lymphotoxin-alpha (LT-α) or tumor necrosis factor-beta (TNF-β) is a protein that in humans is encoded by the LTA gene . (wikidoc.org)
  • TNF-beta, or lymphotoxin, is produced by lymphocytes. (medscape.com)
  • Activation of the receptor by CD70 ANTIGEN results in the increased proliferation of CD4-POSITIVE T-LYMPHOCYTES and CD8-POSITIVE T-LYMPHOCYTES. (bvsalud.org)
  • NF-kappaB-inducing kinase is dispensable for activation of NF-kappaB in inflammatory settings but essential for lymphotoxin beta receptor activation of NF-kappaB in primary human fibroblasts. (ox.ac.uk)
  • After LT-β receptor activation, IKK-α, β, and γ are produced, which increases degradation of I-κB , an inhibitor or NF-kB, and produce NF-kB1 (p50) and ReIA (p60). (wikidoc.org)
  • Activation of LT-β receptors is capable of inducing cell death of cancerous cells and suppressing tumor growth. (wikidoc.org)
  • Enhancement of hepatitis B virus replication by androgen and its receptor in mice J Virol. (usc.edu)
  • Absence of lymph nodes in lymphotoxin-alpha (LTalpha)-deficient mice is due to abnormal organ development, not defective lymphocyte migration, Journal of Inflammation , Vol.45 , (号) , 72-78, 1995. (tokushima-u.ac.jp)
  • Affinity maturation without germinal centres in lymphotoxin-alpha-deficient mice, Nature , Vol.382 , No.6590 , 462-466, 1996. (tokushima-u.ac.jp)
  • Lymphotoxin-alpha deficient and TNF receptor-I-deficient mice define developmental and functional characteristics of germinal centers, Immunological Reviews , Vol.156 , (号) , 137-144, (month) 1997. (tokushima-u.ac.jp)
  • The canonical pathway is activated by signals from various immune-related receptors. (xiahepublishing.com)
  • Lymphotoxin beta receptor has been shown to interact with Diablo homolog and TRAF3. (wikipedia.org)
  • LT-α 1 -β 2 can interact with receptors such as LT-β receptors . (wikidoc.org)
  • LT and TNF belong to the same family, interact with a common receptor, and could act together during disease progression to effect the pathogenesis, according to recent evidence. (plos.org)
  • The thymic selection of the human T-cell receptor repertoire releases polyspecific receptors with the ability to recognize and respond to peptides from unrelated viruses. (elifesciences.org)
  • Although initially identified by co-expression of conventional αβ T-cell receptors (TCR) and markers typically associated with natural killer (NK) cells [ 3 ], NKT are currently distinguished on the basis of CD1d restriction as well as specific usage of TCRα chains [ 4 ]. (biomedcentral.com)
  • However, TNF-RI functions as the high affinity receptor for soluble TNF (sTNF). (hycultbiotech.com)
  • Antibodies against SARS-CoV-2 spike receptor-binding domain were quantified in serum. (bvsalud.org)
  • Does not cross-react with TNF-beta, lymphotoxin and receptor bound TNF-alpha. (lsbio.com)
  • It does not react with receptor-bound TNF-alpha. (abcam.com)
  • Description: A sandwich quantitative ELISA assay kit for detection of Mouse Lymphotoxin Beta (LTb) in samples from serum, plasma, tissue homogenates, cell lysates or other biological fluids. (hudsen.org)
  • Description: This is Double-antibody Sandwich Enzyme-linked immunosorbent assay for detection of Mouse Lymphotoxin Beta (LTb) in serum, plasma, tissue homogenates, cell lysates and other biological fluids. (hudsen.org)
  • Description: Enzyme-linked immunosorbent assay based on the Double-antibody Sandwich method for detection of Mouse Lymphotoxin Beta (LTb) in samples from serum, plasma, tissue homogenates, cell lysates and other biological fluids with no significant corss-reactivity with analogues from other species. (hudsen.org)
  • Description: A sandwich ELISA kit for detection of Lymphotoxin Beta from Mouse in samples from blood, serum, plasma, cell culture fluid and other biological fluids. (hudsen.org)
  • Effect of gene polymorphism of TNF-beta on the concentration of TNF in serum of patient with endometriosis]. (cdc.gov)
  • Tumor necrosis factor (TNF)-TNF receptor gene polymorphisms and their serum levels in Korean women with endometriosis. (cdc.gov)
  • Association between endometriosis and polymorphisms in tumor necrosis factor-related apoptosis-inducing ligand (TRAIL), TRAIL receptor and osteoprotegerin genes and their serum levels. (cdc.gov)
  • alpha- and beta-interferon crossreact with common receptors, while gamma-interferon initiates its biological effects through its own specific receptor system. (lookformedical.com)
  • The interferon alpha-beta receptor signals through the action of JANUS KINASES such as the TYK2 KINASE. (lookformedical.com)
  • Upon their secretion by antigen-presenting cells, they exert both pro-inflammatory and anti-inflammatory receptor-mediated effects. (nature.com)
  • The alpha and beta angles are 90 degrees while the gamma angle is 120 degrees. (wikipedia.org)
  • LT-α can also form heterotrimers with lymphotoxin-beta , which anchors lymphotoxin-alpha to the cell surface. (wikidoc.org)
  • They include alpha- and beta-interferons ( INTERFERON-ALPHA and INTERFERON-BETA ). (lookformedical.com)
  • A ubiquitously expressed heterodimeric receptor that is specific for both INTERFERON-ALPHA and INTERFERON-BETA . (lookformedical.com)
  • Distinct roles of lymphotoxin-alpha and the type I TNF receptor in the establishment of follicular dendritic cells from non-bone marrow-derived cells, The Journal of Experimental Medicine , Vol.186 , No.12 , 1997-2004, 1997. (tokushima-u.ac.jp)
  • Lymphotoxin-alpha (LTalpha) supports development of splenic follicular structure that is required for IgG responses, The Journal of Experimental Medicine , Vol.185 , No.12 , 2111-2120, 1997. (tokushima-u.ac.jp)
  • Genetic contribution of tumor necrosis factor (TNF)-alpha gene promoter (-1031, -863 and -857) and TNF receptor 2 gene polymorphisms in endometriosis susceptibility. (cdc.gov)
  • sis factor alpha (TNF-), tumor necrosis factor beta (TNF- ), interleukin (IL)-1, and IL-2 (11-14). (cdc.gov)
  • Role of lymphotoxin and the type I TNF receptor in the formation of germinal centers, Science , Vol.271 , No.5253 , 1289-1291, 1996. (tokushima-u.ac.jp)
  • In experimental cerebral malaria, TNF-beta, now called lymphotoxin α (LT), has been shown to be a principal mediator of pathogenesis. (plos.org)