Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Cell Rep ; 38(12): 110553, 2022 03 22.
Artículo en Inglés | MEDLINE | ID: mdl-35320716

RESUMEN

The Btla inhibitory receptor limits innate and adaptive immune responses, both preventing the development of autoimmune disease and restraining anti-viral and anti-tumor responses. It remains unclear how the functions of Btla in diverse lymphocytes contribute to immunoregulation. Here, we show that Btla inhibits activation of genes regulating metabolism and cytokine signaling, including Il6 and Hif1a, indicating a regulatory role in humoral immunity. Within mucosal Peyer's patches, we find T-cell-expressed Btla-regulated Tfh cells, while Btla in T or B cells regulates GC B cell numbers. Treg-expressed Btla is required for cell-intrinsic Treg homeostasis that subsequently controls GC B cells. Loss of Btla in lymphocytes results in increased IgA bound to intestinal bacteria, correlating with altered microbial homeostasis and elevations in commensal and pathogenic bacteria. Together our studies provide important insights into how Btla functions as a checkpoint in diverse conventional and regulatory lymphocyte subsets to influence systemic immune responses.


Asunto(s)
Inmunidad Humoral , Linfocitos T Reguladores , Linfocitos B , Mucosa Intestinal , Transducción de Señal
2.
Front Immunol ; 11: 1268, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32676079

RESUMEN

Neuronal regulation of diverse physiological functions requires complex molecular interactions in innervated tissues to maintain proper organ function. Here we show that loss of the neuronal cell surface adhesion/recognition molecule Contactin-1 (Cntn1) directly impairs intestinal function causing wasting that subsequently results in global immune defects. Loss of Cntn1 results in hematologic alterations and changes in blood metabolites associated with malnourishment. We found thymus and spleen of Cntn1-deficient animals atrophied with severe reductions in lymphocyte populations. Elevated thymic Gilz expression indicated ongoing glucocorticoid signaling in Cntn1-deficient animals, consistent with the malnourishment phenotype. Intestinal Contactin-1 was localized to neurons in the villi and the submucosal/myenteric plexus that innervates smooth muscle. Loss of Cntn1 was associated with reduced intestinal Bdnf and Adrb2, indicating reduced neuromuscular crosstalk. Additionally, loss of Cntn1 resulted in reduced recruitment of CD3+ T cells to villi within the small intestine. Together, these data illustrate the critical role of Contactin-1 function within the gut, and how this is required for normal systemic immune functions.


Asunto(s)
Contactina 1/genética , Mucosa Intestinal/inmunología , Mucosa Intestinal/inervación , Animales , Biomarcadores , Recuento de Células Sanguíneas , Análisis Químico de la Sangre , Citometría de Flujo , Perfilación de la Expresión Génica , Glucocorticoides/metabolismo , Homeostasis , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patología , Ratones , Ratones Noqueados , Fenotipo , Transducción de Señal , Bazo/inmunología , Bazo/metabolismo , Bazo/patología , Timo/inmunología , Timo/metabolismo , Timo/patología
3.
Adv Cancer Res ; 142: 145-186, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30885361

RESUMEN

Somatic mutations in cancer cells may influence tumor growth, survival, or immune interactions in their microenvironment. The tumor necrosis factor receptor family member HVEM (TNFRSF14) is frequently mutated in cancers and has been attributed a tumor suppressive role in some cancer contexts. HVEM functions both as a ligand for the lymphocyte checkpoint proteins BTLA and CD160, and as a receptor that activates NF-κB signaling pathways in response to BTLA and CD160 and the TNF ligands LIGHT and LTα. BTLA functions to inhibit lymphocyte activation, but has also been ascribed a role in stimulating cell survival. CD160 functions to co-stimulate lymphocyte function, but has also been shown to activate inhibitory signaling in CD4+ T cells. Thus, the role of HVEM within diverse cancers and in regulating the immune responses to these tumors is likely context specific. Additionally, development of therapeutics that target proteins within this network of interacting proteins will require a deeper understanding of how these proteins function in a cancer-specific manner. However, the prominent role of the HVEM network in anti-cancer immune responses indicates a promising area for drug development.


Asunto(s)
Neoplasias/genética , Neoplasias/inmunología , Miembro 14 de Receptores del Factor de Necrosis Tumoral/genética , Miembro 14 de Receptores del Factor de Necrosis Tumoral/inmunología , Linfocitos T/inmunología , Genes Supresores de Tumor , Humanos , Activación de Linfocitos , Mutación , Subunidad p50 de NF-kappa B/inmunología , Subunidad p50 de NF-kappa B/metabolismo , Neoplasias/metabolismo , Neoplasias/patología , Miembro 14 de Receptores del Factor de Necrosis Tumoral/metabolismo , Transducción de Señal/genética , Transducción de Señal/inmunología , Microambiente Tumoral
4.
J Immunol ; 202(7): 2057-2068, 2019 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-30770415

RESUMEN

CD160 is highly expressed by NK cells and is associated with cytolytic effector activity. Herpes virus entry mediator (HVEM) activates NK cells for cytokine production and cytolytic function via CD160. Fc-fusions are a well-established class of therapeutics, where the Fc domain provides additional biological and pharmacological properties to the fusion protein including enhanced serum t 1/2 and interaction with Fc receptor-expressing immune cells. We evaluated the specific function of HVEM in regulating CD160-mediated NK cell effector function by generating a fusion of the HVEM extracellular domain with human IgG1 Fc bearing CD16-binding mutations (Fc*) resulting in HVEM-(Fc*). HVEM-(Fc*) displayed reduced binding to the Fc receptor CD16 (i.e., Fc-disabled HVEM), which limited Fc receptor-induced responses. HVEM-(Fc*) functional activity was compared with HVEM-Fc containing the wild type human IgG1 Fc. HVEM-(Fc*) treatment of NK cells and PBMCs caused greater IFN-γ production, enhanced cytotoxicity, reduced NK fratricide, and no change in CD16 expression on human NK cells compared with HVEM-Fc. HVEM-(Fc*) treatment of monocytes or PBMCs enhanced the expression level of CD80, CD83, and CD40 expression on monocytes. HVEM-(Fc*)-enhanced NK cell activation and cytotoxicity were promoted via cross-talk between NK cells and monocytes that was driven by cell-cell contact. In this study, we have shown that soluble Fc-disabled HVEM-(Fc*) augments NK cell activation, IFN-γ production, and cytotoxicity of NK cells without inducing NK cell fratricide by promoting cross-talk between NK cells and monocytes without Fc receptor-induced effects. Soluble Fc-disabled HVEM-(Fc*) may be considered as a research and potentially therapeutic reagent for modulating immune responses via sole activation of HVEM receptors.


Asunto(s)
Células Asesinas Naturales/inmunología , Activación de Linfocitos/inmunología , Monocitos/inmunología , Miembro 14 de Receptores del Factor de Necrosis Tumoral/inmunología , Antígenos CD/inmunología , Células Cultivadas , Proteínas Ligadas a GPI/inmunología , Humanos , Fragmentos Fc de Inmunoglobulinas/inmunología , Inmunoglobulina G/inmunología , Receptor Cross-Talk , Receptores Inmunológicos/inmunología , Transducción de Señal/inmunología
5.
J Biol Chem ; 292(51): 21060-21070, 2017 12 22.
Artículo en Inglés | MEDLINE | ID: mdl-29061848

RESUMEN

The human cytomegalovirus opening reading frame UL144 is an ortholog of the TNF receptor superfamily member, herpesvirus entry mediator (HVEM; TNFRSF14). HVEM binds the TNF ligands, LIGHT and LTa; the immunoglobulin inhibitory receptor, B and T lymphocyte attenuator (BTLA); and the natural killer cell-activating receptor CD160. However, UL144 selectively binds BTLA, avoiding activation of inflammatory signaling initiated by CD160 in natural killer cells. BTLA and CD160 cross-compete for binding HVEM, but the structural basis for the ligand selectivity by UL144 and how it acts as an anti-inflammatory agonist remains unclear. Here, we modeled the UL144 structure and characterized its binding with BTLA. The UL144 structure was predicted to closely mimic the surface of HVEM, and we also found that both HVEM and UL144 bind a common epitope of BTLA, whether engaged in trans or in cis, that is shared with a BTLA antibody agonist. On the basis of the UL144 selectivity, we engineered a BTLA-selective HVEM protein to understand the basis for ligand selectivity and BTLA agonism to develop novel anti-inflammatory agonists. This HVEM mutein did not bind CD160 or TNF ligands but did bind BTLA with 10-fold stronger affinity than wild-type HVEM and retained potent inhibitory activity that reduced T-cell receptor, B-cell receptor, and interferon signaling in B cells. In conclusion, using a viral immune evasion strategy that shows broad immune-ablating activity, we have identified a novel anti-inflammatory BTLA-selective agonist.


Asunto(s)
Linfocitos B/metabolismo , Células Asesinas Naturales/metabolismo , Glicoproteínas de Membrana/metabolismo , Modelos Moleculares , Receptores Inmunológicos/agonistas , Miembro 14 de Receptores del Factor de Necrosis Tumoral/metabolismo , Linfocitos T/metabolismo , Proteínas Virales/metabolismo , Sustitución de Aminoácidos , Antiinflamatorios no Esteroideos/química , Antiinflamatorios no Esteroideos/metabolismo , Antiinflamatorios no Esteroideos/farmacología , Antígenos CD/química , Antígenos CD/genética , Antígenos CD/metabolismo , Linfocitos B/citología , Linfocitos B/efectos de los fármacos , Linfocitos B/inmunología , Sitios de Unión , Línea Celular Tumoral , Diseño de Fármacos , Proteínas Ligadas a GPI/química , Proteínas Ligadas a GPI/genética , Proteínas Ligadas a GPI/metabolismo , Células HEK293 , Humanos , Células Asesinas Naturales/citología , Células Asesinas Naturales/efectos de los fármacos , Células Asesinas Naturales/inmunología , Cinética , Ligandos , Glicoproteínas de Membrana/química , Glicoproteínas de Membrana/genética , Mutación , Conformación Proteica , Ingeniería de Proteínas , Dominios y Motivos de Interacción de Proteínas , Receptores Inmunológicos/química , Receptores Inmunológicos/genética , Receptores Inmunológicos/metabolismo , Miembro 14 de Receptores del Factor de Necrosis Tumoral/química , Miembro 14 de Receptores del Factor de Necrosis Tumoral/genética , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/metabolismo , Proteínas Recombinantes de Fusión/farmacología , Linfocitos T/citología , Linfocitos T/efectos de los fármacos , Linfocitos T/inmunología , Proteínas Virales/química , Proteínas Virales/genética
6.
Immunity ; 44(5): 1005-19, 2016 05 17.
Artículo en Inglés | MEDLINE | ID: mdl-27192566

RESUMEN

Cytokines related to tumor necrosis factor (TNF) provide a communication network essential for coordinating multiple cell types into an effective host defense system against pathogens and malignant cells. The pathways controlled by the TNF superfamily differentiate both innate and adaptive immune cells and modulate stromal cells into microenvironments conducive to host defenses. Members of the TNF receptor superfamily activate diverse cellular functions from the production of type 1 interferons to the modulation of survival of antigen-activated T cells. Here, we focus attention on the subset of TNF superfamily receptors encoded in the immune response locus in chromosomal region 1p36. Recent studies have revealed that these receptors use diverse mechanisms to either co-stimulate or restrict immune responses. Translation of the fundamental mechanisms of TNF superfamily is leading to the design of therapeutics that can alter pathogenic processes in several autoimmune diseases or promote immunity to tumors.


Asunto(s)
Enfermedades Autoinmunes/inmunología , Trastornos de los Cromosomas/genética , Inmunoterapia/métodos , Neoplasias/inmunología , Receptor Cross-Talk , Receptores del Factor de Necrosis Tumoral/metabolismo , Linfocitos T/inmunología , Inmunidad Adaptativa , Animales , Deleción Cromosómica , Cromosomas Humanos Par 1/genética , Humanos , Inmunidad Innata , Inmunoterapia/tendencias , Activación de Linfocitos , Neurogénesis/genética , Transducción de Señal
7.
Hepatology ; 63(5): 1576-91, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-26799785

RESUMEN

UNLABELLED: The hypoxia-inducible factor (HIF), HIF-1, is a central regulator of the response to low oxygen or inflammatory stress and plays an essential role in survival and function of immune cells. However, the mechanisms regulating nonhypoxic induction of HIF-1 remain unclear. Here, we assess the impact of germline heterozygosity of a novel, oxygen-independent ubiquitin ligase for HIF-1α: hypoxia-associated factor (HAF; encoded by SART1). SART1(-/-) mice were embryonic lethal, whereas male SART1(+/-) mice spontaneously recapitulated key features of nonalcoholic steatohepatitis (NASH)-driven hepatocellular carcinoma (HCC), including steatosis, fibrosis, and inflammatory cytokine production. Male, but not female, SART1(+/-) mice showed significant up-regulation of HIF-1α in circulating and liver-infiltrating immune cells, but not in hepatocytes, before development of malignancy. Additionally, Kupffer cells derived from male, but not female, SART1(+/-) mice produced increased levels of the HIF-1-dependent chemokine, regulated on activation, normal T-cell expressed and secreted (RANTES), compared to wild type. This was associated with increased liver-neutrophilic infiltration, whereas infiltration of lymphocytes and macrophages were not significantly different. Neutralization of circulating RANTES decreased liver neutrophilic infiltration and attenuated HCC tumor initiation/growth in SART1(+/-) mice. CONCLUSION: This work establishes a new tumor-suppressor role for HAF in immune cell function by preventing inappropriate HIF-1 activation in male mice and identifies RANTES as a novel therapeutic target for NASH and NASH-driven HCC.


Asunto(s)
Carcinoma Hepatocelular/etiología , Quimiocina CCL5/fisiología , Haploinsuficiencia , Subunidad alfa del Factor 1 Inducible por Hipoxia/fisiología , Neoplasias Hepáticas/etiología , Transactivadores/genética , Animales , Ácidos Grasos/metabolismo , Hígado Graso/etiología , Femenino , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Infiltración Neutrófila , Ribonucleoproteínas Nucleares Pequeñas
8.
Cold Spring Harb Perspect Biol ; 7(4): a016279, 2014 Dec 18.
Artículo en Inglés | MEDLINE | ID: mdl-25524549

RESUMEN

The tumor necrosis factor superfamily (TNFSF) and its corresponding receptor superfamily (TNFRSF) form communication pathways required for developmental, homeostatic, and stimulus-responsive processes in vivo. Although this receptor-ligand system operates between many different cell types and organ systems, many of these proteins play specific roles in immune system function. The TNFSF and TNFRSF proteins lymphotoxins, LIGHT (homologous to lymphotoxins, exhibits inducible expression, and competes with HSV glycoprotein D for herpes virus entry mediator [HVEM], a receptor expressed by T lymphocytes), lymphotoxin-ß receptor (LT-ßR), and HVEM are used by embryonic and adult innate lymphocytes to promote the development and homeostasis of lymphoid organs. Lymphotoxin-expressing innate-acting B cells construct microenvironments in lymphoid organs that restrict pathogen spread and initiate interferon defenses. Recent results illustrate how the communication networks formed among these cytokines and the coreceptors B and T lymphocyte attenuator (BTLA) and CD160 both inhibit and activate innate lymphoid cells (ILCs), innate γδ T cells, and natural killer (NK) cells. Understanding the role of TNFSF/TNFRSF and interacting proteins in innate cells will likely reveal avenues for future therapeutics for human disease.


Asunto(s)
Inmunidad Innata , Inflamación/inmunología , Linfocitos/metabolismo , Receptores del Factor de Necrosis Tumoral/metabolismo , Factores de Necrosis Tumoral/metabolismo , Animales , Antígenos CD/metabolismo , Proteínas Ligadas a GPI/metabolismo , Homeostasis , Humanos , Inflamación/metabolismo , Heterotrímero de Linfotoxina alfa1 y beta2/metabolismo , Receptores Inmunológicos/metabolismo
9.
Front Immunol ; 5: 654, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25566265

RESUMEN

Lymphocytes of the gamma delta (γδ) T-cell lineage are evolutionary conserved and although they express rearranged antigen-specific receptors, a large proportion respond as innate effectors. γδ T-cells are poised to combat infection by responding rapidly to cytokine stimuli similar to innate lymphoid cells. This potential to initiate strong inflammatory responses necessitates that inhibitory signals are balanced with activation signals. Here, we discuss some of the key mechanisms that regulate the development, activation, and inhibition of innate γδ T-cells in light of recent evidence that the inhibitory immunoglobulin-superfamily member B and T lymphocyte attenuator restricts their differentiation and effector function.

10.
Immunity ; 39(6): 1082-1094, 2013 Dec 12.
Artículo en Inglés | MEDLINE | ID: mdl-24315996

RESUMEN

γδ T cells rapidly secrete inflammatory cytokines at barrier sites that aid in protection from pathogens, but mechanisms limiting inflammatory damage remain unclear. We found that retinoid-related orphan receptor gamma-t (RORγt) and interleukin-7 (IL-7) influence γδ T cell homeostasis and function by regulating expression of the inhibitory receptor, B and T lymphocyte attenuator (BTLA). The transcription factor RORγt, via its activating function-2 domain, repressed Btla transcription, whereas IL-7 increased BTLA levels on the cell surface. BTLA expression limited γδ T cell numbers and sustained normal γδ T cell subset frequencies by restricting IL-7 responsiveness and expansion of the CD27(-)RORγt(+) population. BTLA also negatively regulated IL-17 and TNF production in CD27(-) γδ T cells. Consequently, BTLA-deficient mice exhibit enhanced disease in a γδ T cell-dependent model of dermatitis, whereas BTLA agonism reduced inflammation. Therefore, by coordinating expression of BTLA, RORγt and IL-7 balance suppressive and activation stimuli to regulate γδ T cell homeostasis and inflammatory responses.


Asunto(s)
Homeostasis , Inflamación , Receptores Inmunológicos/metabolismo , Linfocitos T/metabolismo , Animales , Células Cultivadas , Modelos Animales de Enfermedad , Citometría de Flujo , Eliminación de Gen , Humanos , Ratones , Ratones Endogámicos C57BL , Receptores de Antígenos de Linfocitos T gamma-delta/inmunología , Receptores Inmunológicos/genética , Linfocitos T/inmunología
11.
J Immunol ; 191(9): 4611-8, 2013 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-24078690

RESUMEN

Innate lymphoid cells encompass a diverse array of lymphocyte subsets with unique phenotype that initiate inflammation and provide host defenses in specific microenvironments. In this study, we identify a rare human CD4(+)CD3(-) innate-like lymphoid population with high TNF expression that is enriched in blood from patients with rheumatoid arthritis. These CD4(+)CD3(-) cells belong to the T cell lineage, but the lack of AgR at the cell surface renders them nonresponsive to TCR-directed stimuli. By developing a culture system that sustains survival, we show that CD4(+)CD3(-) innate-like T cells display IL-7-dependent induction of surface lymphotoxin-αß, demonstrating their potential to modify tissue microenvironments. Furthermore, expression of CCR6 on the CD4(+)CD3(-) population defines a CD127(high) subset that is highly responsive to IL-7. This CD4(+)CD3(-) population is enriched in the peripheral blood from rheumatoid arthritis patients, suggesting a link to their involvement in chronic inflammatory disease.


Asunto(s)
Artritis Reumatoide/metabolismo , Complejo CD3/metabolismo , Linfocitos T CD4-Positivos/metabolismo , Heterotrímero de Linfotoxina alfa1 y beta2/metabolismo , Factores de Necrosis Tumoral/metabolismo , Adulto , Anciano , Animales , Artritis Reumatoide/inmunología , Linfocitos T CD4-Positivos/inmunología , Línea Celular , Femenino , Células HEK293 , Proteínas de Homeodominio/genética , Humanos , Inflamación , Interleucina-7/metabolismo , Subunidad alfa del Receptor de Interleucina-7/metabolismo , Activación de Linfocitos/inmunología , Recuento de Linfocitos , Heterotrímero de Linfotoxina alfa1 y beta2/inmunología , Masculino , Ratones , Ratones Noqueados , Persona de Mediana Edad , Receptores CCR6/metabolismo
12.
J Immunol ; 191(2): 828-36, 2013 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-23761635

RESUMEN

Lymphocyte activation is regulated by costimulatory and inhibitory receptors, of which both B and T lymphocyte attenuator (BTLA) and CD160 engage herpesvirus entry mediator (HVEM). Notably, it remains unclear how HVEM functions with each of its ligands during immune responses. In this study, we show that HVEM specifically activates CD160 on effector NK cells challenged with virus-infected cells. Human CD56(dim) NK cells were costimulated specifically by HVEM but not by other receptors that share the HVEM ligands LIGHT, Lymphotoxin-α, or BTLA. HVEM enhanced human NK cell activation by type I IFN and IL-2, resulting in increased IFN-γ and TNF-α secretion, and tumor cell-expressed HVEM activated CD160 in a human NK cell line, causing rapid hyperphosphorylation of serine kinases ERK1/2 and AKT and enhanced cytolysis of target cells. In contrast, HVEM activation of BTLA reduced cytolysis of target cells. Together, our results demonstrate that HVEM functions as a regulator of immune function that activates NK cells via CD160 and limits lymphocyte-induced inflammation via association with BTLA.


Asunto(s)
Antígenos CD/metabolismo , Células Asesinas Naturales/inmunología , Receptores Inmunológicos/metabolismo , Miembro 14 de Receptores del Factor de Necrosis Tumoral/metabolismo , Antígeno CD56/metabolismo , Línea Celular , Activación Enzimática , Proteínas Ligadas a GPI/metabolismo , Células HEK293 , Humanos , Inflamación , Interferón Tipo I/metabolismo , Interferón gamma/metabolismo , Interleucina-2/metabolismo , Células Asesinas Naturales/metabolismo , Activación de Linfocitos , Linfotoxina-alfa/metabolismo , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Fosforilación , Proteínas Proto-Oncogénicas c-akt/metabolismo , Miembro 14 de Receptores del Factor de Necrosis Tumoral/inmunología , Transducción de Señal , Linfocitos T/inmunología , Miembro 14 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo
13.
Curr Opin Immunol ; 23(5): 627-31, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21920726

RESUMEN

The herpesvirus entry mediator (HVEM; TNFRSF14) can activate either proinflammatory or inhibitory signaling pathways. HVEM engages two distinct types of ligands, the canonical TNF-related cytokines, LIGHT and Lymphotoxin-α, and the Ig-related membrane proteins, BTLA (B and T lymphocyte attenuator) and CD160. Recent evidence indicates that the signal generated by HVEM depends on the context of its ligands expressed in trans or in cis. HVEM engagement by all of its ligands in trans initiates bidirectional signaling. In contrast, naïve T cells coexpress BTLA and HVEM forming a cis-complex that interferes with the activation of HVEM by extraneous ligands in the surrounding microenvironment. The HVEM Network is emerging as a key survival system for effector and memory T cells in mucosal tissues.


Asunto(s)
Herpes Simple/inmunología , Inmunidad Innata , Membrana Mucosa/inmunología , Receptores Inmunológicos/inmunología , Miembro 14 de Receptores del Factor de Necrosis Tumoral/inmunología , Transducción de Señal/inmunología , Simplexvirus/inmunología , Linfocitos T/inmunología , Animales , Antígenos CD/inmunología , Antígenos CD/metabolismo , Microambiente Celular , Proteínas Ligadas a GPI/inmunología , Proteínas Ligadas a GPI/metabolismo , Herpes Simple/virología , Humanos , Evasión Inmune , Ligandos , Activación de Linfocitos/inmunología , Linfotoxina-alfa/inmunología , Linfotoxina-alfa/metabolismo , Ratones , Ratones Noqueados , Membrana Mucosa/citología , Membrana Mucosa/metabolismo , Membrana Mucosa/virología , Receptores Inmunológicos/metabolismo , Miembro 14 de Receptores del Factor de Necrosis Tumoral/metabolismo , Linfocitos T/citología , Linfocitos T/metabolismo , Linfocitos T/virología
14.
Nat Rev Immunol ; 8(11): 861-73, 2008 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-18949019

RESUMEN

Herpesviruses have evolved numerous strategies to subvert host immune responses so they can coexist with their host species. These viruses 'co-opt' host genes for entry into host cells and then express immunomodulatory genes, including mimics of members of the tumour-necrosis factor (TNF) superfamily, that initiate and alter host-cell signalling pathways. TNF superfamily members have crucial roles in controlling herpesvirus infection by mediating the direct killing of infected cells and by enhancing immune responses. Despite these strong immune responses, herpesviruses persist in a latent form, which suggests a dynamic relationship between the host immune system and the virus that results in a balance between host survival and viral control.


Asunto(s)
Infecciones por Herpesviridae/inmunología , Herpesviridae/inmunología , Receptores del Factor de Necrosis Tumoral/inmunología , Factores de Necrosis Tumoral/inmunología , Animales , Infecciones por Herpesviridae/virología , Humanos
15.
PLoS One ; 3(7): e2823, 2008 Jul 30.
Artículo en Inglés | MEDLINE | ID: mdl-18665263

RESUMEN

Recent reports suggested that Delta1, 4 and Jagged1, 2 possessed the ability to instruct CD4(+) T cell into selection of Th1 or Th2 fates, respectively, although the underlying mechanism endowing the cleaved Notch receptor with memory of ligand involved in its activation remains elusive. To examine this, we prepared artificial antigen-presenting cells expressing either DLL1 or Jag1. Although both ligands were efficient in inducing Notch2 cleavage and activation in CD4(+) T or reporter cells, the presence of Lunatic Fringe in CD4(+) T cells inhibited Jag1 activation of Notch1 receptor. Neither ligand could induce Th1 or Th2 fate choice independently of cytokines or redirect cytokine-driven Th1 or Th2 development. Instead, we find that Notch ligands only augment cytokine production during T cell differentiation in the presence of polarizing IL-12 and IL-4. Moreover, the differentiation choices of naïve CD4(+) T cells lacking gamma-secretase, RBP-J, or both in response to polarizing cytokines revealed that neither presenilin proteins nor RBP-J were required for cytokine-induced Th1/Th2 fate selection. However, presenilins facilitate cellular proliferation and cytokine secretion in an RBP-J (and thus, Notch) independent manner. The controversies surrounding the role of Notch and presenilins in Th1/Th2 polarization may reflect their role as genetic modifiers of T-helper cells differentiation.


Asunto(s)
Citocinas/metabolismo , Presenilinas/metabolismo , Receptores Notch/metabolismo , Células TH1/metabolismo , Células Th2/metabolismo , Animales , Células CHO , Diferenciación Celular , Linaje de la Célula , Cricetinae , Cricetulus , Interleucina-12/metabolismo , Interleucina-4/metabolismo , Ratones , Ratones Endogámicos C57BL
16.
J Immunol ; 180(2): 940-7, 2008 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-18178834

RESUMEN

The B and T lymphocyte attenuator (BTLA) appears to act as a negative regulator of T cell activation and growth. BTLA specifically interacts with herpesvirus entry mediator (HVEM), a member of the TNFR family. Herein, we have undertaken surface plasmon resonance studies to quantitatively assess BTLA and HVEM ectodomain interactions. We find that soluble BALB/cJ BTLA engages HVEM with an equilibrium affinity of 0.97+/-0.19 microM while the C57BL/6 BTLA binds slightly better with an equilibrium affinity of 0.42+/-0.06 microM. Despite its lower affinity for HVEM, the kinetic half-life of BALB/cJ BTLA complexes are twice as long as observed for C57BL/6 BTLA (4 vs 2 s). To further explore these interactions, we solved the crystal structure of a murine BTLA (BALB/cJ) ectodomain at 1.8-A resolution, revealing a beta sandwich fold with strong similarity to I-set members of the Ig superfamily. Using a structure-based mutagenesis strategy, we then examined the individual contributions of 26 BTLA surface-exposed residues toward HVEM binding. Four single-site substitutions were identified that decrease HVEM binding below detectable levels and two that decrease binding by more than half. All six of these cluster at the edge of the beta sandwich in a membrane distal patch formed primarily from the A and G strands. This patch falls within the contacting surface recently revealed in the crystal structure of the human BTLA-HVEM cocomplex. The critical binding residues identified here are highly conserved across species, suggesting that BTLA employs a conserved binding mode for HVEM recognition.


Asunto(s)
Receptores Inmunológicos/química , Miembro 14 de Receptores del Factor de Necrosis Tumoral/química , Secuencia de Aminoácidos , Sustitución de Aminoácidos , Animales , Secuencia Conservada , Cristalografía por Rayos X , Análisis Mutacional de ADN , Perros , Humanos , Ratones , Datos de Secuencia Molecular , Conformación Proteica , Mapeo de Interacción de Proteínas , Ratas , Receptores Inmunológicos/genética , Resonancia por Plasmón de Superficie
17.
J Immunol ; 178(10): 6073-82, 2007 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-17475832

RESUMEN

B and T lymphocyte attenuator (BTLA; CD272) can deliver inhibitory signals to B and T cells upon binding its ligand herpesvirus entry mediator. Because CD28, CTLA-4, programmed death-1, and ICOS regulate the development of acute graft-vs-host disease (GVHD), we wished to assess if BTLA also played a role in this T cell-mediated response. In the nonirradiated parental-into-F1 model of acute GVHD, BTLA+/+ and BTLA-/- donor lymphocytes showed equivalent engraftment and expansion during the first week of the alloresponse. Unexpectedly, BTLA-/- donor T cells failed to sustain GVHD, showing a decline in surviving donor cell numbers beginning at day 9 and greatly reduced by day 11. Similarly, inhibition of BTLA-herpesvirus entry mediator engagement by in vivo administration of a blocking anti-BTLA Ab also caused reduced survival of donor cells. Microarray analysis revealed several genes that were differentially expressed by BTLA-/- and BTLA+/+ donor CD4+ T cells preceding the decline in BTLA-/- donor T cells. Several genes influencing Th cell polarization were differentially expressed by BTLA+/+ and BTLA-/- donor cells. Additionally, the re-expression of the IL-7Ralpha subunit that occurs in BTLA+/+ donor cells after 1 wk of in vivo allostimulation was not observed in BTLA-/- donor CD4+ cells. The striking loss of BTLA-/- T cells in this model indicates a role for BTLA activity in sustaining CD4+ T cell survival under the conditions of chronic stimulation in the nonirradiated parental-into-F1 GVHD.


Asunto(s)
Subgrupos de Linfocitos B/citología , Subgrupos de Linfocitos B/inmunología , Isoantígenos/inmunología , Activación de Linfocitos/inmunología , Receptores Inmunológicos/fisiología , Subgrupos de Linfocitos T/citología , Subgrupos de Linfocitos T/inmunología , Animales , Anticuerpos Monoclonales/administración & dosificación , Supervivencia Celular/inmunología , Enfermedad Crónica , Modelos Animales de Enfermedad , Supervivencia de Injerto/inmunología , Enfermedad Injerto contra Huésped/inmunología , Enfermedad Injerto contra Huésped/metabolismo , Enfermedad Injerto contra Huésped/patología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Endogámicos MRL lpr , Ratones Noqueados , Receptores Inmunológicos/deficiencia , Receptores Inmunológicos/genética , Receptores Inmunológicos/inmunología
18.
Int Immunol ; 19(6): 763-73, 2007 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-17513880

RESUMEN

The T cell Ig domain and mucin domain (TIM) proteins form a conserved family of transmembrane cell-surface glycoproteins expressed by a variety of tissues. Each TIM protein contains a single V-type Ig domain, a glycosylated mucin-like domain, a transmembrane domain and a cytoplasmic domain. TIM proteins recognize a diverse array of ligands, including H-ferritin, galectin-9 as well as other TIM family members. In this study, we demonstrate that the Ig domains of murine TIM-1, -3 and -4 display calcium-dependent binding to ligands expressed by murine splenocytes and several non-murine cell lines, indicating non-species-specific ligand recognition. Further, the intrafamilial interaction of various TIM family Ig domains with surface-expressed TIM-1 and TIM-4 requires an intact TIM-1 and TIM-4 glycosylated mucin stalk. Importantly, we also uncovered the previously unrecognized potential for homotypic TIM interactions in forming ligand-receptor pairs. Using a glycan array screen, we identified the novel capacity of the TIM-3 Ig domain to recognize specific carbohydrate moieties, suggesting a role for carbohydrate modification along with protein epitopes in TIM ligand recognition. Identification of the carbohydrate-binding capacity of TIM proteins helps explain the diversity of ligands recognized by this family and adds to our understanding of homotypic and heterotypic interactions between TIM family members.


Asunto(s)
Proteínas de la Membrana/metabolismo , Polisacáridos/metabolismo , Receptores Virales/metabolismo , Animales , Células CHO , Calcio/metabolismo , Calcio/farmacología , Línea Celular , Sulfatos de Condroitina/metabolismo , Cricetinae , Cricetulus , Ácido Egtácico/farmacología , Heparitina Sulfato/metabolismo , Receptor Celular 1 del Virus de la Hepatitis A , Receptor 2 Celular del Virus de la Hepatitis A , Leucocitos Mononucleares/metabolismo , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Proteínas de la Membrana/genética , Ratones , Ratones Endogámicos BALB C , Fragmentos de Péptidos/genética , Fragmentos de Péptidos/metabolismo , Polisacáridos/química , Unión Proteica/efectos de los fármacos , Receptores Virales/genética , Proteínas Recombinantes de Fusión/metabolismo , Bazo/citología , Spodoptera , Transfección
19.
Adv Immunol ; 92: 157-85, 2006.
Artículo en Inglés | MEDLINE | ID: mdl-17145304

RESUMEN

Recently a new inhibitory immunoglobulin domain-containing lymphocyte receptor was identified on the basis of its T helper 1 (T(H)1)-selective expression in murine T cell lines, which was named B and T lymphocyte attenuator (BTLA). Several groups have confirmed the initial characterization of BTLA as an inhibitory receptor, which was initially inferred from the mild increases in several parameters of BTLA-deficient mice. The initial expectation that BTLA would interact with a B7 family ligand, such as the B7x protein, was surprisingly overturned with the functional cloning of the actual BTLA ligand as herpesvirus entry mediator (HVEM). This was unexpected largely due to the fact that this interaction represents the convergence of two very different, although each quite extensive, families of receptors and ligands. The interaction of BTLA, which belongs to the CD28 family of the immunoglobulin superfamily, and HVEM, a costimulatory tumor-necrosis factor (TNF) receptor (TNFR), is quite unique in that it is the only receptor-ligand interaction that directly bridges these two families of receptors. This interaction has raised many questions about how receptors from two different families could interact and which are the signaling events downstream of receptor ligation. As we discuss here and recently demonstrated, HVEM interaction with BTLA serves to negatively regulate T cell responses, in contrast to the strong activation observed when HVEM engages its endogenous ligand from the TNF family. Finally, as studies of BTLA are just now beginning to extend beyond the initial characterizations, it is becoming clear that there are many complex issues remaining to be resolved, particularly potential polymorphisms that may engender disease susceptibility in the human.


Asunto(s)
Células Presentadoras de Antígenos/inmunología , Activación de Linfocitos/inmunología , Receptor Cross-Talk/fisiología , Receptores Inmunológicos/fisiología , Miembro 14 de Receptores del Factor de Necrosis Tumoral/fisiología , Linfocitos T/inmunología , Secuencia de Aminoácidos , Animales , Humanos , Datos de Secuencia Molecular , Receptor Cross-Talk/inmunología
20.
Nat Rev Immunol ; 6(9): 671-81, 2006 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-16932752

RESUMEN

The interaction between B- and T-lymphocyte attenuator (BTLA), an inhibitory receptor whose extracellular domain belongs to the immunoglobulin superfamily, and herpesvirus-entry mediator (HVEM), a co-stimulatory tumour-necrosis factor receptor, is unique in that it is the only receptor-ligand interaction that directly bridges these two families of receptors. This interaction has raised many questions about how receptors from two different families could interact and what downstream signalling events might occur as a result of receptor ligation. As we discuss, recent studies show that engagement of HVEM with its endogenous ligand (LIGHT) from the tumour-necrosis factor family induces a powerful immune response, whereas HVEM interactions with BTLA negatively regulate T-cell responses.


Asunto(s)
Receptores Inmunológicos/inmunología , Receptores del Factor de Necrosis Tumoral/inmunología , Receptores Virales/inmunología , Animales , Enfermedad , Humanos , Ligandos , Unión Proteica , Receptores Inmunológicos/antagonistas & inhibidores , Receptores Inmunológicos/química , Receptores del Factor de Necrosis Tumoral/antagonistas & inhibidores , Receptores del Factor de Necrosis Tumoral/química , Receptores del Factor de Necrosis Tumoral/clasificación , Miembro 14 de Receptores del Factor de Necrosis Tumoral , Receptores Virales/antagonistas & inhibidores , Receptores Virales/química , Receptores Virales/clasificación , Transducción de Señal
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA