Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Ann Rheum Dis ; 2024 May 03.
Artículo en Inglés | MEDLINE | ID: mdl-38569851

RESUMEN

INTRODUCTION: Anifrolumab is a type I interferon (IFN) receptor 1 (IFNAR1) blocking antibody approved for treating patients with systemic lupus erythematosus (SLE). Here, we investigated the immunomodulatory mechanisms of anifrolumab using longitudinal transcriptomic and proteomic analyses of the 52-week, randomised, phase 3 TULIP-1 and TULIP-2 trials. METHODS: Patients with moderate to severe SLE were enrolled in TULIP-1 and TULIP-2 and received intravenous anifrolumab or placebo alongside standard therapy. Whole-blood expression of 18 017 genes using genome-wide RNA sequencing (RNA-seq) (pooled TULIP; anifrolumab, n=244; placebo, n=258) and 184 plasma proteins using Olink and Simoa panels (TULIP-1; anifrolumab, n=124; placebo, n=132) were analysed. We compared treatment groups via gene set enrichment analysis using MetaBase pathway analysis, blood transcriptome modules, in silico deconvolution of RNA-seq and longitudinal linear mixed effect models for gene counts and protein levels. RESULTS: Compared with placebo, anifrolumab modulated >2000 genes by week 24, with overlapping results at week 52 and 41 proteins by week 52. IFNAR1 blockade with anifrolumab downregulated multiple type I and II IFN-induced gene modules/pathways and type III IFN-λ protein levels, and impacted apoptosis-associated and neutrophil extracellular trap-associated transcriptional pathways, innate cell activating chemokines and receptors, proinflammatory cytokines and B-cell activating cytokines. In silico deconvolution of RNA-seq data indicated an increase from baseline of mucosal-associated invariant and γδT cells and a decrease of monocytes following anifrolumab treatment. DISCUSSION: Type I IFN blockade with anifrolumab modulated multiple inflammatory pathways downstream of type I IFN signalling, including apoptotic, innate and adaptive mechanisms that play key roles in SLE immunopathogenesis.

2.
Cancer Immunol Res ; 10(2): 200-214, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34937728

RESUMEN

Checkpoint blockade therapies targeting PD-1/PD-L1 and CTLA-4 are clinically successful but also evoke adverse events due to systemic T-cell activation. We engineered a bispecific, mAb targeting CD28 homolog (CD28H), a newly identified B7 family receptor that is constitutively expressed on T and natural killer (NK) cells, with a PD-L1 antibody to potentiate tumor-specific immune responses. The bispecific antibody led to T-cell costimulation, induced NK-cell cytotoxicity of PD-L1-expressing tumor cells, and activated tissue-resident memory CD8+ T cells. Mechanistically, the CD28H agonistic arm of the bispecific antibody reduced PD-L1/PD-1-induced SHP2 phosphorylation while simultaneously augmenting T-cell receptor signaling by activating the MAPK and AKT pathways. This bispecific approach could be used to target multiple immune cells, including CD8+ T cells, tissue-resident memory T cells, and NK cells, in a tumor-specific manner that may lead to induction of durable, therapeutic antitumor responses.


Asunto(s)
Anticuerpos Biespecíficos , Neoplasias , Anticuerpos Biespecíficos/metabolismo , Antígeno B7-H1/metabolismo , Antígenos CD28/metabolismo , Linfocitos T CD8-positivos , Línea Celular Tumoral , Humanos , Inmunoterapia , Células Asesinas Naturales , Activación de Linfocitos , Neoplasias/metabolismo , Receptor de Muerte Celular Programada 1/metabolismo
3.
Int J Mol Sci ; 22(20)2021 Oct 19.
Artículo en Inglés | MEDLINE | ID: mdl-34681945

RESUMEN

Therapeutic success in treating patients with systemic lupus erythematosus (SLE) is limited by the multivariate disease etiology, multi-organ presentation, systemic involvement, and complex immunopathogenesis. Agents targeting B-cell differentiation and survival are not efficacious for all patients, indicating a need to target other inflammatory mediators. One such target is the type I interferon pathway. Type I interferons upregulate interferon gene signatures and mediate critical antiviral responses. Dysregulated type I interferon signaling is detectable in many patients with SLE and other autoimmune diseases, and the extent of this dysregulation is associated with disease severity, making type I interferons therapeutically tangible targets. The recent approval of the type I interferon-blocking antibody, anifrolumab, by the US Food and Drug Administration for the treatment of patients with SLE demonstrates the value of targeting this pathway. Nevertheless, the interferon pathway has pleiotropic biology, with multiple cellular targets and signaling components that are incompletely understood. Deconvoluting the complexity of the type I interferon pathway and its intersection with lupus disease pathology will be valuable for further development of targeted SLE therapeutics. This review summarizes the immune mediators of the interferon pathway, its association with disease pathogenesis, and therapeutic modalities targeting the dysregulated interferon pathway.


Asunto(s)
Antivirales/farmacología , Enfermedades Autoinmunes/tratamiento farmacológico , Inmunidad Innata/efectos de los fármacos , Interferón Tipo I/farmacología , Lupus Eritematoso Sistémico/tratamiento farmacológico , Animales , Enfermedades Autoinmunes/patología , Humanos , Lupus Eritematoso Sistémico/patología , Transducción de Señal
4.
Sci Transl Med ; 10(431)2018 03 07.
Artículo en Inglés | MEDLINE | ID: mdl-29514998

RESUMEN

Systemic sclerosis (SSc) is a debilitating inflammatory and fibrotic disease that affects the skin and internal organs. Although the pathophysiology of SSc remains poorly characterized, mononuclear cells, mainly macrophages and T cells, have been implicated in inflammation and fibrosis. Inducible costimulator (ICOS), which is expressed on a subset of memory T helper (TH) and T follicular helper (TFH) cells, has been shown to be increased in SSc and associated with disease pathology. However, the identity of the relevant ICOS+ T cells and their contribution to inflammation and fibrosis in SSc are still unknown. We show that CD4+ ICOS-expressing T cells with a TFH-like phenotype infiltrate the skin of patients with SSc and are correlated with dermal fibrosis and clinical disease status. ICOS+ TFH-like cells were found to be increased in the skin of graft-versus-host disease (GVHD)-SSc mice and contributed to dermal fibrosis via an interleukin-21- and matrix metalloproteinase 12-dependent mechanism. Administration of an anti-ICOS antibody to GVHD-SSc mice prevented the expansion of ICOS+ TFH-like cells and inhibited inflammation and dermal fibrosis. Interleukin-21 neutralization in GVHD-SSc mice blocked disease pathogenesis by reducing skin fibrosis. These results identify ICOS+ TFH-like profibrotic cells as key drivers of fibrosis in a GVHD-SSc model and suggest that inhibition of these cells could offer therapeutic benefit for SSc.


Asunto(s)
Fibrosis/inmunología , Fibrosis/metabolismo , Esclerodermia Sistémica/inmunología , Esclerodermia Sistémica/metabolismo , Linfocitos T/metabolismo , Animales , Femenino , Fibrosis/terapia , Enfermedad Injerto contra Huésped/inmunología , Enfermedad Injerto contra Huésped/metabolismo , Enfermedad Injerto contra Huésped/terapia , Humanos , Proteína Coestimuladora de Linfocitos T Inducibles/metabolismo , Interleucinas/antagonistas & inhibidores , Interleucinas/metabolismo , Ratones , Ratones Endogámicos BALB C , Receptores de Interleucina-21/metabolismo , Esclerodermia Sistémica/terapia , Piel/inmunología , Piel/metabolismo , Enfermedades de la Piel/inmunología , Enfermedades de la Piel/metabolismo , Enfermedades de la Piel/terapia
5.
Nat Commun ; 7: 13895, 2016 12 23.
Artículo en Inglés | MEDLINE | ID: mdl-28008916

RESUMEN

Mutations affecting the apoptosis-inducing function of the Fas/CD95 TNF-family receptor result in autoimmune and lymphoproliferative disease. However, Fas can also costimulate T-cell activation and promote tumour cell growth and metastasis. Palmitoylation at a membrane proximal cysteine residue enables Fas to localize to lipid raft microdomains and induce apoptosis in cell lines. Here, we show that a palmitoylation-defective Fas C194V mutant is defective in inducing apoptosis in primary mouse T cells, B cells and dendritic cells, while retaining the ability to enhance naive T-cell differentiation. Despite inability to efficiently induce cell death, the Fas C194V receptor prevents the lymphoaccumulation and autoimmunity that develops in Fas-deficient mice. These findings indicate that induction of apoptosis through Fas is dependent on receptor palmitoylation in primary immune cells, and Fas may prevent autoimmunity by mechanisms other than inducing apoptosis.


Asunto(s)
Apoptosis/inmunología , Autoinmunidad/inmunología , Microdominios de Membrana/inmunología , Ratones Transgénicos , Receptor fas/inmunología , Animales , Apoptosis/genética , Autoinmunidad/genética , Diferenciación Celular/genética , Diferenciación Celular/inmunología , Células Cultivadas , Células HEK293 , Humanos , Lipoilación/inmunología , Microdominios de Membrana/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados , Mutación , Linfocitos T/inmunología , Linfocitos T/metabolismo , Receptor fas/genética , Receptor fas/metabolismo
6.
J Clin Invest ; 126(1): 318-34, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26657860

RESUMEN

Adoptive cell transfer (ACT) of purified naive, stem cell memory, and central memory T cell subsets results in superior persistence and antitumor immunity compared with ACT of populations containing more-differentiated effector memory and effector T cells. Despite a clear advantage of the less-differentiated populations, the majority of ACT trials utilize unfractionated T cell subsets. Here, we have challenged the notion that the mere presence of less-differentiated T cells in starting populations used to generate therapeutic T cells is sufficient to convey their desirable attributes. Using both mouse and human cells, we identified a T cell-T cell interaction whereby antigen-experienced subsets directly promote the phenotypic, functional, and metabolic differentiation of naive T cells. This process led to the loss of less-differentiated T cell subsets and resulted in impaired cellular persistence and tumor regression in mouse models following ACT. The T memory-induced conversion of naive T cells was mediated by a nonapoptotic Fas signal, resulting in Akt-driven cellular differentiation. Thus, induction of Fas signaling enhanced T cell differentiation and impaired antitumor immunity, while Fas signaling blockade preserved the antitumor efficacy of naive cells within mixed populations. These findings reveal that T cell subsets can synchronize their differentiation state in a process similar to quorum sensing in unicellular organisms and suggest that disruption of this quorum-like behavior among T cells has potential to enhance T cell-based immunotherapies.


Asunto(s)
Memoria Inmunológica , Inmunoterapia Adoptiva , Linfocitos T/inmunología , Animales , Diferenciación Celular , Proteína Ligando Fas/fisiología , Femenino , Melanoma Experimental/terapia , Ratones , Ratones Endogámicos C57BL , Proteínas Proto-Oncogénicas c-akt/fisiología , Linfocitos T/citología , Receptor fas/fisiología
7.
J Immunol ; 193(9): 4303-11, 2014 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-25246497

RESUMEN

Bcl-3 is an atypical member of the IκB family and modulates gene expression via interaction with p50/NF-κB1 or p52/NF-κB2 homodimers. We report in the present study that Bcl-3 is required in dendritic cells (DCs) to assure effective priming of CD4 and CD8 T cells. Lack of Bcl-3 in bone marrow-derived DCs blunted their ability to expand and promote effector functions of T cells upon Ag/adjuvant challenge in vitro and after adoptive transfers in vivo. Importantly, the critical role of Bcl-3 for priming of T cells was exposed upon Ag/adjuvant challenge of mice specifically ablated of Bcl-3 in DCs. Furthermore, Bcl-3 in endogenous DCs was necessary for contact hypersensitivity responses. Bcl-3 modestly aided maturation of DCs, but most consequentially, Bcl-3 promoted their survival, partially inhibiting expression of several antiapoptotic genes. Loss of Bcl-3 accelerated apoptosis of bone marrow-derived DCs during Ag presentation to T cells, and DC survival was markedly impaired in the context of inflammatory conditions in mice specifically lacking Bcl-3 in these cells. Conversely, selective overexpression of Bcl-3 in DCs extended their lifespan in vitro and in vivo, correlating with increased capacity to prime T cells. These results expose a previously unidentified function for Bcl-3 in DC survival and the generation of adaptive immunity.


Asunto(s)
Presentación de Antígeno , Células Dendríticas/inmunología , FN-kappa B/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Factores de Transcripción/metabolismo , Animales , Proteínas del Linfoma 3 de Células B , Antígeno CD11c/metabolismo , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Diferenciación Celular , Supervivencia Celular/genética , Células Dendríticas/citología , Células Dendríticas/metabolismo , Expresión Génica , Activación de Linfocitos/inmunología , Ratones , Ratones Noqueados , Proteínas Proto-Oncogénicas/genética , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo , Factores de Transcripción/genética
8.
Methods Enzymol ; 542: 125-49, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24862264

RESUMEN

Evidence accumulating over the past decade has linked alterations in bioenergetic metabolism to the pathogenesis of several diseases, including inflammatory conditions and cancer. However, the mutual relationship between the effector functions and the metabolism of immune cells has begun to emerge only recently. Similar to malignant cells, both innate and adaptive immune cells undergo a metabolic reprogramming that is required for effector functions, de facto underlying the elicitation of a robust immune response. These changes allow immune cells not only to rapidly respond to pathogens or (pre)malignant cells but also to adapt to changing microenvironmental conditions. Targeting the metabolic alterations of malignant cells has been the subject of an intense wave of investigation, resulting in the identification of promising therapeutic strategies. Since the inflammatory milieu and the tumor microenvironment are similar, the metabolism of immune cells and its regulation has recently come under renewed interest as a target for immunotherapy. Here, we describe different tools and techniques to study the bioenergetic metabolism of cultured cells, using immune cells as a model. Our methodological approach relies on an extracellular flux analyzer, an instrument that enables the real-time measurement of the two central pathways used by living cells to generate adenosine triphosphate: glycolysis and oxidative phosphorylation. This instrument and similar technological innovations have transformed the study of cellular metabolism, unveiling its profound impact on various immunologic and oncological disorders.


Asunto(s)
Bioquímica/métodos , Glucólisis , Sistema Inmunológico/citología , Mitocondrias/metabolismo , Oxígeno/metabolismo , Bioquímica/instrumentación , Células Cultivadas , Metabolismo Energético , Espacio Extracelular/metabolismo , Humanos , Sistema Inmunológico/metabolismo , Células Jurkat , Linfocitos/metabolismo , Metaboloma , Células Mieloides/metabolismo , Fosforilación Oxidativa , Análisis de Matrices Tisulares
9.
Methods Mol Biol ; 979: 33-41, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23397386

RESUMEN

Among the different techniques available for determining physiological cell death or apoptosis in immune cells, fluorescence-activated cell sorting-based approaches prove to be one of the most efficient and quantitative assays in capturing cells that are actively undergoing apoptosis elicited by either extrinsic or intrinsic forms of cell death. The key advantage of the technique is to allow the user to determine apoptotic responses of multiple immune cell types or subsets of the same lineage of immune cells without the prior requirement for cell separation. Here, we describe a "multiparameter" flow method to rapidly determine apoptosis-sensitivity induced by the Fas receptor pathway within different CD4 T cell subsets in a mixed pool of analyzed ex vivo peripheral blood mononuclear cells.


Asunto(s)
Apoptosis , Citometría de Flujo/métodos , Subgrupos de Linfocitos T/citología , Linfocitos T CD4-Positivos/citología , Linfocitos T CD4-Positivos/metabolismo , Ficoll/química , Humanos , Subgrupos de Linfocitos T/metabolismo , Receptor fas/metabolismo
10.
J Clin Immunol ; 33(2): 479-88, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23054345

RESUMEN

Deleterious mutations in genes involved in the Fas apoptosis pathway lead to Autoimmune Lymphoproliferative Syndrome (ALPS). Demonstration of an apoptosis defect is critical for the diagnosis and study of ALPS. The traditional in vitro apoptosis assay, however, requires a week of experimental procedures. Here, we show that defects in Fas-induced apoptosis in PBMCs can be evaluated directly ex vivo using multicolor flow cytometry to analyze the apoptosis of effector memory T cells, a Fas-sensitive subset of PBMCs. This method allowed us to sensitively quantify defective apoptosis in ALPS patients within a few hours. Some ALPS patients (ALPS-sFAS) without germline mutations have somatic mutations in Fas specifically in double-negative αß T cells (DNTs), an unusual lymphocyte population that is characteristically expanded in ALPS. Since DNTs have been notoriously difficult to culture, defective apoptosis has not been previously demonstrated for ALPS-sFAS patients. Using our novel ex vivo apoptosis assay, we measured Fas-induced apoptosis of DNTs for the first time and found that ALPS-sFAS patients had significant apoptosis defects in these cells compared to healthy controls. Hence, this rapid apoptosis assay can expedite the diagnosis of new ALPS patients, including those with somatic mutations, and facilitate clinical and molecular investigation of these diseases.


Asunto(s)
Apoptosis , Síndrome Linfoproliferativo Autoinmune/diagnóstico , Pruebas Inmunológicas de Citotoxicidad/métodos , Linfocitos T/metabolismo , Receptor fas/metabolismo , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Apoptosis/inmunología , Síndrome Linfoproliferativo Autoinmune/genética , Síndrome Linfoproliferativo Autoinmune/inmunología , Niño , Preescolar , Citometría de Flujo , Humanos , Persona de Mediana Edad , Mutación , Reproducibilidad de los Resultados , Sensibilidad y Especificidad , Linfocitos T/inmunología , Adulto Joven , Receptor fas/genética
12.
Cell Immunol ; 271(2): 210-3, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21903206

RESUMEN

Th1 cells are remarkably more susceptible to activation induced cell death than Th17. Here, we compared cultures of these two cell subpopulations for their expression of apoptosis-related molecules when re-exposed to their specific antigen. We also compared the expression of apoptosis-related molecules in the mouse eye with inflammation induced by Th1 or Th17 cells. Using qPCR we found that the mRNA transcript levels of the majority of tested apoptosis-related molecules were higher in the Th1 cultures, and in eyes with Th1-induced inflammation. Apoptotic intrinsic pathway molecules played minor roles in the processes in vitro or in vivo, whereas extrinsic pathway molecules, as well as PD-1, its ligands and Tim3, were heavily involved.


Asunto(s)
Apoptosis/inmunología , Células TH1/inmunología , Células TH1/metabolismo , Células Th17/inmunología , Células Th17/metabolismo , Traslado Adoptivo , Animales , Línea Celular , Oftalmopatías/etiología , Oftalmopatías/inmunología , Inflamación/etiología , Inflamación/inmunología , Activación de Linfocitos , Ratones , Ratones Transgénicos
13.
Nat Rev Rheumatol ; 7(3): 152-60, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21283145

RESUMEN

Programmed cell death (PCD) is a key process in the regulation of immune cell development and peripheral immune homeostasis. Caspase-dependent apoptosis, as well as a number of alternative cell death mechanisms, account for immune cell PCD induced by cell-intrinsic and extrinsic pathways. In animal models, compelling evidence has emerged that genetic defects in PCD can result in autoimmune disease. Autoimmune disease can arise from single-gene mutations that affect PCD, and defective PCD has been observed in some tissues and cells from patients with rheumatic disease. Selectively inducing PCD in autoreactive B and T cells is very attractive as a therapeutic strategy because it offers the possibility of permanent elimination of these pathogenic cell subsets. In addition, the anti-inflammatory effects of apoptotic cells may add to the therapeutic benefit of induced PCD. Immune cell subsets vary widely in their sensitivity to specific inducers of cell death, and understanding these differences is key to predicting the outcome of inducing apoptosis for therapeutic means. Here, we review approaches that have been used to induce PCD in the treatment of autoimmune disease, and describe the prospects of bringing these experimental strategies into clinical practice.


Asunto(s)
Apoptosis/fisiología , Enfermedades Reumáticas/tratamiento farmacológico , Enfermedades Reumáticas/fisiopatología , Animales , Enfermedades Autoinmunes/tratamiento farmacológico , Enfermedades Autoinmunes/fisiopatología , Caspasas/fisiología , Humanos , Modelos Animales
14.
J Immunol ; 183(11): 7547-56, 2009 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-19890052

RESUMEN

Both Th1 and Th17 T cell subsets can mediate inflammation, but the kinetics of the pathogenic processes mediated by these two subsets have not been investigated. Using an experimental system in which TCR-transgenic Th1 or Th17 cells specific for hen egg lysozyme induce ocular inflammation in recipient mice expressing eye-restricted hen egg lysozyme, we found important differences in the in vivo behavior of these two subsets. Th1 cells initially proliferated considerably faster and invaded the eye more quickly than their Th17 counterparts, but then disappeared rapidly. By contrast, Th17 cells accumulated and remained the majority of the infiltrating CD4(+) cells in the eye for as long as 25 days after transfer, mediating more long-lasting pathological changes. Unlike Th1, Th17 cells were highly resistant to restimulation-induced apoptosis, a major pathway by which autoimmune and chronically restimulated Th1 cells are eliminated. Th17 cells had reduced Fas ligand production and resistance to Fas-induced apoptosis, relative to Th1 cells, despite similar surface expression of Fas. Th17-induced ocular inflammation also differed from Th1-induced inflammation by consisting of more neutrophils, whereas Th1-induced disease had higher proportions of CD8 cells. Taken together, our data show that pathogenic processes triggered by Th17 lag behind those induced by Th1, but then persist remarkably longer, apparently due to the relative resistance of Th17 cells to restimulation-induced cell death. The long-lasting inflammation induced by Th17 cells is in accord with these cells being involved in chronic conditions in humans.


Asunto(s)
Apoptosis/inmunología , Enfermedades Autoinmunes/inmunología , Linfocitos T CD4-Positivos/inmunología , Inflamación/inmunología , Subgrupos de Linfocitos T/inmunología , Traslado Adoptivo , Animales , Linfocitos T CD8-positivos , División Celular , Ojo/inmunología , Proteína Ligando Fas/inmunología , Proteína Ligando Fas/metabolismo , Citometría de Flujo , Interleucina-17/biosíntesis , Interleucina-17/inmunología , Ratones , Ratones Transgénicos , Muramidasa/inmunología , Células TH1/inmunología , Receptor fas/inmunología , Receptor fas/metabolismo
15.
Results Probl Cell Differ ; 49: 17-47, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19132321

RESUMEN

Interactions between the TNF-family receptor Fas (CD95) and Fas Ligand (FasL, CD178) can efficiently induce apoptosis and are critical for the maintenance of immunological self-tolerance. FasL is kept under strict control by transcriptional and posttranslational regulation. Surface FasL can be cleaved by metalloproteases, resulting in shed extracellular domains, and FasL can also traffic to secretory lysosomes. Each form of FasL has distinct biological functions. Fas is more ubiquitously expressed, but its apoptosis-inducing function is regulated by a number of mechanisms including submembrane localization, efficiency of receptor signaling complex assembly and activation, and bcl-2 family members in some circumstances. When apoptosis is not induced, Fas-FasL interactions can also trigger a number of activating and proinflammatory signals. Harnessing the apoptosis-inducing potential of Fas for therapy of cancer and autoimmune disease has been actively pursued, and despite a number of unexpected side-effects that result from manipulating Fas-FasL interactions, this remains a worthy goal.


Asunto(s)
Apoptosis/inmunología , Proteína Ligando Fas/inmunología , Transducción de Señal/inmunología , Receptor fas/inmunología , Animales , Apoptosis/efectos de los fármacos , Enfermedades Autoinmunes/tratamiento farmacológico , Enfermedades Autoinmunes/inmunología , Proteína Ligando Fas/biosíntesis , Humanos , Inflamación/inmunología , Inflamación/metabolismo , Neoplasias/tratamiento farmacológico , Neoplasias/inmunología , Estructura Terciaria de Proteína , Proteínas Proto-Oncogénicas c-bcl-2/inmunología , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Receptor fas/biosíntesis , Receptor fas/uso terapéutico
16.
J Immunol ; 179(10): 6384-8, 2007 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-17982024

RESUMEN

In activated CD4(+) T cells, TCR restimulation triggers apoptosis that depends on interactions between the death receptor Fas and its ligand, FasL. This process, termed restimulation-induced cell death (RICD), is a mechanism of peripheral immune tolerance. TCR signaling sensitizes activated T cells to Fas-mediated apoptosis, but what pathways mediate this process are not known. In this study we identify the Rho GTPases Rac1 and Rac2 as essential components in restimulation-induced cell death. RNA interference-mediated knockdown of Rac GTPases greatly reduced Fas-dependent, TCR-induced apoptosis. The ability of Rac1 to sensitize T cells to Fas-induced apoptosis correlated with Rac-mediated cytoskeletal reorganization, dephosphorylation of the ERM (ezrin/radixin/moesin) family of cytoskeletal linker proteins, and the translocation of Fas to lipid raft microdomains. In primary activated CD4(+) T cells, Rac1 and Rac2 were independently required for maximal TCR-induced apoptosis. Activating Rac signaling may be a novel way to sensitize chronically stimulated lymphocytes to Fas-induced apoptosis, an important goal in the treatment of autoimmune diseases.


Asunto(s)
Apoptosis/inmunología , Linfocitos T CD4-Positivos/inmunología , Tolerancia Inmunológica , Activación de Linfocitos/inmunología , Receptor fas/inmunología , Proteínas de Unión al GTP rac/inmunología , Proteína de Unión al GTP rac1/inmunología , Enfermedades Autoinmunes/enzimología , Enfermedades Autoinmunes/inmunología , Enfermedades Autoinmunes/terapia , Linfocitos T CD4-Positivos/citología , Linfocitos T CD4-Positivos/enzimología , Muerte Celular/inmunología , Proteínas del Citoesqueleto/inmunología , Proteínas del Citoesqueleto/metabolismo , Citoesqueleto/inmunología , Citoesqueleto/metabolismo , Activación Enzimática/inmunología , Proteína Ligando Fas , Humanos , Células Jurkat , Microdominios de Membrana/inmunología , Microdominios de Membrana/metabolismo , Proteínas de la Membrana/inmunología , Proteínas de la Membrana/metabolismo , Proteínas de Microfilamentos/inmunología , Proteínas de Microfilamentos/metabolismo , Transporte de Proteínas/inmunología , Receptores de Antígenos de Linfocitos T/inmunología , Receptores de Antígenos de Linfocitos T/metabolismo , Transducción de Señal/inmunología , Receptor fas/metabolismo , Proteínas de Unión al GTP rac/metabolismo , Proteína de Unión al GTP rac1/metabolismo , Proteína RCA2 de Unión a GTP
17.
Immunity ; 26(5): 545-7, 2007 May.
Artículo en Inglés | MEDLINE | ID: mdl-17521581

RESUMEN

In this issue of Immunity,Stranges et al. (2007) ablate expression of the death receptor Fas in T cells, B cells, and DC. Fas deficiency in any of these lineages is sufficient to break self-tolerance.


Asunto(s)
Autotolerancia/inmunología , Receptor fas/inmunología , Animales , Presentación de Antígeno/inmunología , Muerte Celular , Humanos
18.
Oncogene ; 23(36): 6083-94, 2004 Aug 12.
Artículo en Inglés | MEDLINE | ID: mdl-15208670

RESUMEN

Recently, we identified Insulinoma-Glucagonoma clone 20 (IG20) that can render cells more susceptible to tumor necrosis factor-alpha (TNF-alpha)-induced apoptosis. In addition, it can slow cell proliferation, and enhance drug- and radiation-induced cell death. TNF-related apoptosis-inducing ligand (TRAIL) can selectively induce apoptosis in some cancer cells and render others susceptible to cotreatment with drugs and irradiation, with little or no effect on most normal cells. In this study, we investigated the potential of IG20 to enhance TRAIL-induced apoptosis and found that it can render cells more susceptible to TRAIL treatment through enhanced activation of caspases. Further, we showed that this effect can be suppressed by caspase inhibitors, p35 and CrmA, and a dominant-negative Fas-associated death domain-containing protein (DN-FADD). Results from colocalization and immunoprecipitation studies showed that IG20 can interact with TRAIL death receptors (DR), DR4 and DR5 and increase recruitment of FADD and caspase-8 into the TRAIL death-inducing signaling complex (DISC). These results indicate that IG20 is a novel protein that can enhance TRAIL-induced apoptosis by facilitating DISC formation.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Apoptosis , Caspasas/metabolismo , Factores de Intercambio de Guanina Nucleótido/fisiología , Glicoproteínas de Membrana/farmacología , Receptores del Factor de Necrosis Tumoral/metabolismo , Factor de Necrosis Tumoral alfa/farmacología , Apoptosis/efectos de los fármacos , Proteínas Reguladoras de la Apoptosis , Caspasa 8 , Inhibidores de Caspasas , Inhibidores de Cisteína Proteinasa/farmacología , Proteínas Adaptadoras de Señalización del Receptor del Dominio de Muerte , Proteína de Dominio de Muerte Asociada a Fas , Factores de Intercambio de Guanina Nucleótido/genética , Factores de Intercambio de Guanina Nucleótido/metabolismo , Células HeLa , Humanos , Glicoproteínas de Membrana/biosíntesis , Glicoproteínas de Membrana/genética , Complejos Multiproteicos , Mutación , Transporte de Proteínas , Receptores del Ligando Inductor de Apoptosis Relacionado con TNF , Receptores del Factor de Necrosis Tumoral/análisis , Receptores Tipo I de Factores de Necrosis Tumoral/biosíntesis , Receptores Tipo I de Factores de Necrosis Tumoral/genética , Ligando Inductor de Apoptosis Relacionado con TNF , Transcripción Genética , Transfección , Factor de Necrosis Tumoral alfa/biosíntesis , Factor de Necrosis Tumoral alfa/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...