Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Sheng Wu Gong Cheng Xue Bao ; 36(6): 1198-1208, 2020 Jun 25.
Artículo en Chino | MEDLINE | ID: mdl-32597069

RESUMEN

In this research, we studied the formation of Drosophila melanogaster FADD (Fas-associated death domain-containing protein) amyloid fiber and its influence on signal transduction in IMD (Immune deficiency) signaling pathway to better understand the regulation mechanism of Drosophila innate immune signaling pathway, which will provide reference for the immune regulation in other species. First, we purified dFADD protein expressed in Escherichia coli and performed Sulfur flavin T binding and transmission electron microscopy to identify the dFADD amyloid fibers formed in vitro. Then we investigated the formation of dFADD polymers in S2 cells using SDD-AGE and confocal microscope. We also constructed dFADD mutants to find out which domain is essential to fiber formation and its effect on IMD signal transduction. Our results revealed that dFADD could be polymerized to form amyloid fiber polymers in vitro and inside the cells. Formation of fibers relies on DED (Death-effector domain) domain of dFADD, since DED domain-deleted mutant existed as a monomer. Dual luciferase reporter assay showed that intact DED domain was required for the induction of downstream antimicrobial peptides, indicating that fiber formation was the key to IMD signal transduction. Our study revealed the role of dFADD in mediating the cascade between IMD and Dredd in the IMD signaling pathway by forming amyloid fibers, suggesting an evolutionarily conserved regulatory mechanism of innate immune signaling pathway.


Asunto(s)
Proteínas de Drosophila , Drosophila melanogaster , Inmunidad Innata , Transducción de Señal , Animales , Proteínas de Drosophila/biosíntesis , Proteínas de Drosophila/inmunología , Drosophila melanogaster/inmunología , Proteína de Dominio de Muerte Asociada a Fas/biosíntesis , Proteína de Dominio de Muerte Asociada a Fas/inmunología , Inmunidad Innata/inmunología
2.
Fish Shellfish Immunol ; 89: 548-554, 2019 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-30991146

RESUMEN

In this study, a sea cucumber Fas-associated death domain (FADD) named HLFADD was first cloned from Holothuria leucospilota. The full-length cDNA of HLFADD is 2137 bp in size, containing a 116-bp 5'-untranslated region (UTR), a 1334-bp 3'-UTR and a 687-bp open reading frame (ORF) encoding a protein of 228 amino acids with a deduced molecular weight of 26.42 kDa. HLFADD protein contains a conserved death effector domain at its N-terminal and a conserved death domain at its C-terminal, structurally similar to its counterparts in vertebrates. The over-expressed HLFADD protein could induce apoptosis in HEK293 cells, suggesting a possible death receptor-mediated apoptosis pathway in echinoderms adapted with FADD. Moreover, HLFADD mRNA is ubiquitously expressed in all examined tissues, with the highest transcript level in the coelomocytes, followed by intestine. In vitro experiments performed in the H. leucospilota coelomocytes, the expression of HLFADD mRNA was significantly up-regulated by lipopolysaccharides (LPS) or polyriboinosinic-polyribocytidylic acid [poly (I:C)] challenge, suggesting that HLFADD might play important roles in the innate immune defense of sea cucumber against the invasion of bacteria and viruses.


Asunto(s)
Proteína de Dominio de Muerte Asociada a Fas/genética , Proteína de Dominio de Muerte Asociada a Fas/inmunología , Regulación de la Expresión Génica/inmunología , Holothuria/genética , Holothuria/inmunología , Inmunidad Innata/genética , Secuencia de Aminoácidos , Animales , Apoptosis , Secuencia de Bases , Proteína de Dominio de Muerte Asociada a Fas/química , Perfilación de la Expresión Génica , Células HEK293 , Humanos , Lipopolisacáridos/farmacología , Filogenia , Poli I-C/farmacología , Alineación de Secuencia , Regulación hacia Arriba
3.
J Clin Invest ; 129(4): 1551-1565, 2019 02 25.
Artículo en Inglés | MEDLINE | ID: mdl-30694219

RESUMEN

Across clinical trials, T cell expansion and persistence following adoptive cell transfer (ACT) have correlated with superior patient outcomes. Herein, we undertook a pan-cancer analysis to identify actionable ligand-receptor pairs capable of compromising T cell durability following ACT. We discovered that FASLG, the gene encoding the apoptosis-inducing ligand FasL, is overexpressed within the majority of human tumor microenvironments (TMEs). Further, we uncovered that Fas, the receptor for FasL, is highly expressed on patient-derived T cells used for clinical ACT. We hypothesized that a cognate Fas-FasL interaction within the TME might limit both T cell persistence and antitumor efficacy. We discovered that genetic engineering of Fas variants impaired in the ability to bind FADD functioned as dominant negative receptors (DNRs), preventing FasL-induced apoptosis in Fas-competent T cells. T cells coengineered with a Fas DNR and either a T cell receptor or chimeric antigen receptor exhibited enhanced persistence following ACT, resulting in superior antitumor efficacy against established solid and hematologic cancers. Despite increased longevity, Fas DNR-engineered T cells did not undergo aberrant expansion or mediate autoimmunity. Thus, T cell-intrinsic disruption of Fas signaling through genetic engineering represents a potentially universal strategy to enhance ACT efficacy across a broad range of human malignancies.


Asunto(s)
Traslado Adoptivo , Ingeniería Genética , Neoplasias Experimentales/terapia , Receptores Quiméricos de Antígenos , Transducción de Señal/inmunología , Microambiente Tumoral/inmunología , Animales , Proteína Ligando Fas/genética , Proteína Ligando Fas/inmunología , Proteína de Dominio de Muerte Asociada a Fas/genética , Proteína de Dominio de Muerte Asociada a Fas/inmunología , Femenino , Humanos , Masculino , Ratones , Ratones Transgénicos , Neoplasias Experimentales/genética , Neoplasias Experimentales/inmunología , Neoplasias Experimentales/patología , Receptores Quiméricos de Antígenos/genética , Receptores Quiméricos de Antígenos/inmunología , Receptores Quiméricos de Antígenos/uso terapéutico , Transducción de Señal/genética , Microambiente Tumoral/genética , Receptor fas/genética , Receptor fas/inmunología
4.
FEBS J ; 286(3): 523-535, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30536547

RESUMEN

Fas (CD95) signalling is best known for its role in apoptosis, however, recent reports have shown it to be involved in other cellular responses as well, including inflammation. Fas and its adaptor protein FADD are known to negatively regulate LPS-induced proinflammatory responses, but their role in LPS-induced type I interferon production is unknown. Here, we demonstrate that Fas engagement on macrophages, using an agonistic Fas antibody CH11, augments LPS-induced NF-κB responses, causing increased production of TNFα, IL-8, IL-6 and IL-12. Conversely, costimulation with both LPS and CH11 causes a significant reduction in the level of interferon-beta (IFNß) production. This differential effect involves the Fas adaptor FADD because while LPS-induced IL-6 production increased in FADD-/- murine embryonic fibroblasts, LPS-induced IFNß production was significantly reduced in these cells. Overexpression of a dominant negative form of FADD (FADD-DD) inhibits LPS-induced IFNß luciferase but not LPS-induced NF-κB luciferase. In contrast, overexpression of full-length FADD inhibited LPS-induced NF-κB luciferase activation but was seen to augment LPS-induced IFNß luciferase. Moreover, FADD-DD inhibits TRIF-, TRAM-, IKKε-, TBK-1- and TRAF3-induced IFNß luciferase production, with coimmunoprecipitation experiments demonstrating an interaction between FADD and TRIF. These data identify FADD as a novel component of the noncanonical Toll-like receptor 4/IFNß signalling pathway and demonstrate that both Fas and its adaptor FADD can differentially regulate the production of LPS-induced proinflammatory cytokines and type I interferons.


Asunto(s)
Proteína de Dominio de Muerte Asociada a Fas/genética , Interferón beta/genética , Lipopolisacáridos/farmacología , Macrófagos/efectos de los fármacos , Receptor Toll-Like 4/genética , Receptor fas/genética , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/inmunología , Proteínas Adaptadoras del Transporte Vesicular/genética , Proteínas Adaptadoras del Transporte Vesicular/inmunología , Animales , Anticuerpos/farmacología , Proteína de Dominio de Muerte Asociada a Fas/inmunología , Regulación de la Expresión Génica , Células HEK293 , Humanos , Quinasa I-kappa B/genética , Quinasa I-kappa B/inmunología , Interferón beta/inmunología , Interleucina-12/genética , Interleucina-12/inmunología , Interleucina-6/genética , Interleucina-6/inmunología , Interleucina-8/genética , Interleucina-8/inmunología , Células Jurkat , Macrófagos/citología , Macrófagos/inmunología , Ratones , FN-kappa B/genética , FN-kappa B/inmunología , Cultivo Primario de Células , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/inmunología , Células RAW 264.7 , Transducción de Señal , Células THP-1 , Factor 3 Asociado a Receptor de TNF/genética , Factor 3 Asociado a Receptor de TNF/inmunología , Receptor Toll-Like 4/inmunología , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/inmunología , Receptor fas/antagonistas & inhibidores , Receptor fas/inmunología
5.
Trends Immunol ; 39(12): 1036-1053, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30401514

RESUMEN

Initially described as an adaptor molecule for death receptor (DR)-mediated apoptosis, Fas-associated death domain (FADD) was later implicated in nonapoptotic cellular processes. During the last decade, FADD has been shown to participate and regulate most of the signalosome complexes, including necrosome, FADDosome, innateosome, and inflammasome. Given the role of these signaling complexes, FADD has emerged as a new actor in innate immunity, inflammation, and cancer development. Concomitant to these new roles, a surprising number of mechanisms deemed to regulate FADD functions have been identified, including post-translational modifications of FADD protein and FADD secretion. This review focuses on recent knowledge of the biological roles of FADD, a pleiotropic molecule having multiple partners, and its impact in cancer, innate immunity, and inflammation.


Asunto(s)
Proteína de Dominio de Muerte Asociada a Fas/metabolismo , Inflamación/metabolismo , Neoplasias/metabolismo , Animales , Dominio de Muerte , Proteína de Dominio de Muerte Asociada a Fas/inmunología , Humanos , Inflamación/inmunología , Neoplasias/inmunología
6.
Fish Shellfish Immunol ; 74: 517-529, 2018 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-29355760

RESUMEN

Fas-associated protein with death domain (FADD) is the key adaptor protein that transmits apoptotic signals mediated by the main death receptors. Besides being an essential instrument in cell death, FADD is also implicated in proliferation, cell cycle progression, tumor development, inflammation, innate immunity, and autophagy. In the present study, a FADD homologue (EcFADD) from the orange-spotted grouper (Epinephelus coioides) was cloned and its possible role in fish immunity was analyzed. The full length cDNA of EcFADD contains 808 base pairs (bp), including a 573 bp open reading frame that encodes a 190 amino acid protein with a predicted molecular mass of 21.81 kDa. Quantitative real-time polymerase chain reaction analysis indicated that EcFADD was distributed in all examined tissues. The expression of EcFADD in the spleen of E. coioides was differentially up-regulated when challenged with Singapore grouper iridovirus (SGIV) or polyinosine-polycytidylic acid(poly[I:C]). EcFADD was abundantly distributed in both the cytoplasm and nucleus in grouper spleen (GS) and fathead minnow (FHM) epithelial cells. Over-expression of EcFADD inhibited SGIV infection and replication and SGIV-induced apoptosis. To achieve antiviral and anti-apoptosis activities, FADD promoted the activation of interferon-stimulated response element (ISRE) and type I interferon (IFN) genes in the antiviral IFN signaling pathway and inhibited activation of apoptosis-related transcription factors p53. Our results not only characterize FADD but also reveal new immune functions and the molecular mechanisms by which FADD responds to virus infection and virus-induced apoptosis.


Asunto(s)
Lubina/genética , Lubina/inmunología , Proteína de Dominio de Muerte Asociada a Fas/genética , Proteína de Dominio de Muerte Asociada a Fas/inmunología , Enfermedades de los Peces/inmunología , Regulación de la Expresión Génica/inmunología , Inmunidad Innata/genética , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Proteína de Dominio de Muerte Asociada a Fas/química , Proteínas de Peces/química , Proteínas de Peces/genética , Proteínas de Peces/inmunología , Perfilación de la Expresión Génica , Filogenia , Poli I-C/farmacología , Ranavirus/fisiología , Alineación de Secuencia/veterinaria
7.
Biochem Biophys Res Commun ; 485(1): 152-159, 2017 03 25.
Artículo en Inglés | MEDLINE | ID: mdl-28189684

RESUMEN

NF-κB is crucial to regulate immune and inflammatory responses and cell survival. LUBAC generates a linear ubiquitin chain and activates NF-κB through ubiquitin ligase (E3) activity in the HOIP subunit. Here, we show that HOIP is predominantly cleaved by caspase at Asp390 upon apoptosis, and that is subjected to proteasomal degradation. We identified that FADD, as well as NEMO, is a substrate for LUBAC. Although the C-terminal fragment of HOIP retains NF-κB activity, linear ubiquitination of NEMO and FADD decreases upon apoptosis. Moreover, the N-terminal fragment of HOIP binds with deubiquitinases, such as OTULIN and CYLD-SPATA2. These results indicate that caspase-mediated cleavage of HOIP divides critical functional regions of HOIP, and that this regulates linear (de)ubiquitination of substrates upon apoptosis.


Asunto(s)
Apoptosis , Caspasas/metabolismo , Proteína de Dominio de Muerte Asociada a Fas/metabolismo , Quinasa I-kappa B/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitinación , Caspasas/inmunología , Proteína de Dominio de Muerte Asociada a Fas/inmunología , Técnicas de Inactivación de Genes , Células HEK293 , Humanos , Quinasa I-kappa B/inmunología , Células Jurkat , FN-kappa B/inmunología , FN-kappa B/metabolismo , Factor de Necrosis Tumoral alfa/inmunología , Factor de Necrosis Tumoral alfa/metabolismo , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/inmunología
8.
Cell Rep ; 16(12): 3247-3259, 2016 09 20.
Artículo en Inglés | MEDLINE | ID: mdl-27498868

RESUMEN

MLKL, a key component downstream of RIPK3, is suggested to be a terminal executor of necroptosis. Genetic studies have revealed that Ripk3 ablation rescues embryonic lethality in Fadd- or Caspase-8-deficient mice. Given that RIPK3 has also been implicated in non-necroptotic pathways including apoptosis and inflammatory signaling, it remains unclear whether the lethality in Fadd(-/-) mice is indeed caused by necropotosis. Here, we show that genetic deletion of Mlkl rescues the developmental defect in Fadd-deficient mice and that Fadd(-/-)Mlkl(-/-) mice are viable and fertile. Mlkl(-/-)Fadd(-/-) mice display significantly accelerated lymphoproliferative disease characterized by lymphadenopathy and splenomegaly when compared to Ripk3(-/-)Fadd(-/-) mice. Mlkl(-/-)Fadd(-/-) bone-marrow-derived macrophages and dendritic cells have impaired NLRP3 inflammasome activation associated with defects in ASC speck formation and NF-κB-dependent NLRP3 transcription. Our findings reveal that MLKL and FADD play critical roles in preventing lymphoproliferative disease and activating the NLRP3 inflammasome.


Asunto(s)
Proteína de Dominio de Muerte Asociada a Fas/inmunología , Inflamasomas/inmunología , Trastornos Linfoproliferativos/genética , Proteína con Dominio Pirina 3 de la Familia NLR/inmunología , Proteínas Quinasas/inmunología , Animales , Proteína de Dominio de Muerte Asociada a Fas/genética , Trastornos Linfoproliferativos/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Quinasas/genética
9.
J Immunol ; 196(1): 407-15, 2016 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-26582950

RESUMEN

The cytokine IL-1ß is intimately linked to many pathological inflammatory conditions. Mature IL-1ß secretion requires cleavage by the inflammasome. Recent evidence indicates that many cell death signal adaptors have regulatory roles in inflammasome activity. These include the apoptosis inducers FADD and caspase 8, and the necroptosis kinases receptor interacting protein kinase 1 (RIPK1) and RIPK3. PGAM5 is a mitochondrial phosphatase that has been reported to function downstream of RIPK3 to promote necroptosis and IL-1ß secretion. To interrogate the biological function of PGAM5, we generated Pgam5(-/-) mice. We found that Pgam5(-/-) mice were smaller compared with wild type littermates, and male Pgam5(-/-) mice were born at sub-Mendelian ratio. Despite these growth and survival defects, Pgam5(-/-) cells responded normally to multiple inducers of apoptosis and necroptosis. Rather, we found that PGAM5 is critical for IL-1ß secretion in response to NLRP3 and AIM2 inflammasome agonists. Moreover, vesicular stomatosis virus-induced IL-1ß secretion was impaired in Pgam5(-/-) bone marrow-derived macrophages, but not in Ripk3(-/-) bone marrow-derived dendritic cells, indicating that PGAM5 functions independent of RIPK3 to promote inflammasome activation. Mechanistically, PGAM5 promotes ASC polymerization, maintenance of mitochondrial integrity, and optimal reactive oxygen species production in response to inflammasome signals. Hence PGAM5 is a novel regulator of inflammasome and caspase 1 activity that functions independently of RIPK3.


Asunto(s)
Apoptosis/inmunología , Inflamasomas/inmunología , Interleucina-1beta/metabolismo , Macrófagos/inmunología , Monoéster Fosfórico Hidrolasas/genética , Animales , Proteínas Portadoras/inmunología , Caspasa 1/inmunología , Caspasa 8/inmunología , Células Cultivadas , Proteínas de Unión al ADN/inmunología , Células Dendríticas/inmunología , Proteína de Dominio de Muerte Asociada a Fas/inmunología , Inflamación/inmunología , Interleucina-1beta/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mitocondrias/inmunología , Mitocondrias/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR , Fosfoproteínas Fosfatasas , Monoéster Fosfórico Hidrolasas/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Proteína Serina-Treonina Quinasas de Interacción con Receptores/genética , Proteína Serina-Treonina Quinasas de Interacción con Receptores/inmunología , Transducción de Señal/inmunología , Virus de la Estomatitis Vesicular Indiana/inmunología
11.
Nat Rev Immunol ; 14(9): 601-18, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25145756

RESUMEN

Historically, cell death and inflammation have been closely linked, but the necessary divergence of the fields in the past few decades has enriched our molecular understanding of the signalling pathways that mediate various programmes of cell death and multiple types of inflammatory responses. The fields have now come together again demonstrating a surprising level of integration. Intimate interconnections at multiple levels are revealed between the cell death and inflammatory signal transduction pathways that are mobilized in response to the engagement of pattern recognition receptors during microbial infection. Molecules such as receptor-interacting protein kinase 1 (RIPK1), RIPK3, FAS-associated death domain protein (FADD), FLICE-like inhibitory protein (FLIP) and caspase 8 - which are associated with different forms of cell death - are incorporated into compatible and exceedingly dynamic Toll-like receptor, NOD-like receptor and RIG-I-like receptor signalling modules. These signalling modules have a high capacity to switch from inflammation to cell death, or a programmed execution of both, all in an orchestrated battle for host defence and survival.


Asunto(s)
Apoptosis/inmunología , Autofagia/inmunología , Inflamación , Proteínas Adaptadoras del Transporte Vesicular/inmunología , Animales , Proteínas Portadoras/inmunología , Caspasas/genética , Caspasas/inmunología , Proteína de Dominio de Muerte Asociada a Fas/inmunología , Bacterias Gramnegativas/inmunología , Infecciones por Bacterias Gramnegativas/inmunología , Humanos , Ratones , Proteína con Dominio Pirina 3 de la Familia NLR , Proteína Serina-Treonina Quinasas de Interacción con Receptores/inmunología , Transducción de Señal/inmunología , Receptores Toll-Like/inmunología
12.
Curr Opin Pediatr ; 25(6): 722-9, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24240292

RESUMEN

PURPOSE OF REVIEW: Several autoimmune lymphoproliferative syndromes have been described lately. We review here the main clinical and laboratory findings of these new disorders. RECENT FINDINGS: The prototypical autoimmune lymphoproliferative syndrome (ALPS) has had its diagnostic criteria modified, somatic mutations in RAS genes were found to cause an ALPS-like syndrome in humans, and mutations in a gene encoding a protein kinase C (PRKCD) were discovered to cause a syndrome of lymphoproliferation, autoimmunity and natural killer cell defect. SUMMARY: The recent discoveries shed light on the molecular pathways governing lymphocyte death, proliferation and immune tolerance in humans.


Asunto(s)
Apoptosis/inmunología , Síndrome Linfoproliferativo Autoinmune/diagnóstico , Genes ras , Tolerancia Inmunológica/inmunología , Inflamación/inmunología , Apoptosis/genética , Síndrome Linfoproliferativo Autoinmune/genética , Síndrome Linfoproliferativo Autoinmune/inmunología , Autoinmunidad/genética , Proliferación Celular , Proteína de Dominio de Muerte Asociada a Fas/inmunología , Femenino , Genes ras/genética , Genes ras/inmunología , Humanos , Tolerancia Inmunológica/genética , Inflamación/genética , Inflamación/patología , Masculino , Mutación/inmunología , Proteína Quinasa C-delta/inmunología , Transducción de Señal/genética , Transducción de Señal/inmunología
13.
PLoS Pathog ; 9(10): e1003721, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24204270

RESUMEN

RIG-I-Like Receptors (RLRs) sense cytosolic viral RNA to transiently activate type I IFN production. Here, we report that a type I IFN inducible DExD/H helicase, DDX24, exerts a negative-regulatory effect on RLR function. Expression of DDX24 specifically suppressed RLR activity, while DDX24 loss, which caused embryonic lethality, augmented cytosolic RNA-mediated innate signaling and facilitated RNA virus replication. DDX24 preferentially bound to RNA rather than DNA species and influenced signaling by associating with adaptor proteins FADD and RIP1. These events preferentially impeded IRF7 activity, an essential transcription factor for type I IFN production. Our data provide a new function for DDX24 and help explain innate immune gene regulation, mechanisms that may additionally provide insight into the causes of inflammatory disease.


Asunto(s)
ARN Helicasas DEAD-box/inmunología , Inmunidad Innata , ARN Viral/inmunología , Transducción de Señal/inmunología , Estomatitis Vesicular/inmunología , Virus de la Estomatitis Vesicular Indiana/inmunología , Animales , ARN Helicasas DEAD-box/genética , Proteína de Dominio de Muerte Asociada a Fas/genética , Proteína de Dominio de Muerte Asociada a Fas/inmunología , Proteínas Activadoras de GTPasa/genética , Proteínas Activadoras de GTPasa/inmunología , Factor 7 Regulador del Interferón/genética , Factor 7 Regulador del Interferón/inmunología , Interferón Tipo I/genética , Interferón Tipo I/inmunología , Ratones , Ratones Noqueados , ARN Viral/genética , Transducción de Señal/genética , Estomatitis Vesicular/genética , Estomatitis Vesicular/patología , Virus de la Estomatitis Vesicular Indiana/genética
14.
J Immunol ; 188(12): 6194-204, 2012 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-22611243

RESUMEN

We found that an adaptor protein, signal-transducing adaptor protein (STAP)-2, is a new member of the Fas-death-inducing signaling complex and participates in activation-induced cell death in T cells. STAP-2 enhanced Fas-mediated apoptosis and caspase-8 aggregation and activation in Jurkat T cells. Importantly, STAP-2 directly interacted with caspase-8 and Fas, resulting in enhanced interactions between caspase-8 and FADD in the Fas-death-inducing signaling complex. Moreover, STAP-2 protein has a consensus caspase-8 cleavage sequence, VEAD, in its C-terminal domain, and processing of STAP-2 by caspase-8 was crucial for Fas-induced apoptosis. Physiologic roles of STAP-2 were confirmed by observations that STAP-2-deficient mice displayed impaired activation-induced cell death and superantigen-induced T cell depletion. Therefore, STAP-2 is a novel participant in the regulation of T cell apoptosis after stimulation.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Apoptosis/inmunología , Caspasa 8/metabolismo , Proteína de Dominio de Muerte Asociada a Fas/metabolismo , Fosfoproteínas/metabolismo , Linfocitos T/metabolismo , Proteínas Adaptadoras Transductoras de Señales/inmunología , Animales , Western Blotting , Caspasa 8/inmunología , Proteína de Dominio de Muerte Asociada a Fas/inmunología , Citometría de Flujo , Humanos , Inmunoprecipitación , Etiquetado Corte-Fin in Situ , Células Jurkat , Ratones , Ratones Noqueados , Fosfoproteínas/inmunología , ARN Interferente Pequeño , Linfocitos T/inmunología , Transfección , Receptor fas/inmunología , Receptor fas/metabolismo
15.
Rheumatology (Oxford) ; 51(6): 964-75, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22253026

RESUMEN

Inflammation is the principal hallmark of RA. Different pathways are implicated in the production of pro-inflammatory cytokines, the bona fide mediators of this inflammation. Among them are the TNF pathway and the IL-1 receptor/Toll-like receptor (IL-1R/TLR4) pathway. One of the potential negative regulators of IL-1R/TLR4 signalling is the Fas-associated death domain protein (FADD), which is the pivotal adaptor of the apoptotic signal mediated by death receptors of the TNF family. FADD can sequester myeloid differentiation primary response gene 88 (MyD88), the common adaptor of most TLRs, and hence hinder the activation of nuclear factor κB (NF-κB), the downstream transcription factor. We recently described a new regulatory mechanism of FADD expression, via the shedding of microvesicles, mediated by adenosine receptors. Interestingly, adenosine is found in high concentrations in the joints of RA patients and has been largely reported as a regulator of inflammation. This review discusses the possible link that could exist between the adenosine-dependent regulation of FADD in the inflammatory context of RA and the potential role of FADD as a therapeutic target in the treatment of RA. We will see that the modulation of FADD expression may be a double-edged sword by increasing apoptosis and at the same time limiting NF-κB activation.


Asunto(s)
Adenosina/metabolismo , Artritis Reumatoide/inmunología , Artritis Reumatoide/metabolismo , Proteína de Dominio de Muerte Asociada a Fas/inmunología , Proteína de Dominio de Muerte Asociada a Fas/metabolismo , Transducción de Señal/inmunología , Humanos
16.
Hum Immunol ; 73(3): 316-9, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22244917

RESUMEN

An abnormal immune response to common infection(s) may be a plausible etiological mechanism in childhood leukemia. We investigated whether 931 tagging single nucleotide polymorphisms (SNPs) selected in gene regions related to immune response are associated with childhood leukemia susceptibility in a hospital-based case-control study (63 cases and 148 controls) conducted among Korean children. The AT or TT genotype of rs7939734 in Fas-associated protein with death domain (FADD) was associated with increased risk of childhood leukemia compared with the AA genotype (odds ratio [OR] = 2.26, 95% confidence interval [95% CI] = 1.20-4.25, p(trend) = 0.0007, min p = 0.002, false discovery rate [FDR] p = 0.17). The CG or GG genotype of rs2301696 in TRPM5 was associated with decreased risk of childhood leukemia compared with the CC genotype (OR = 0.30, 95% CI = 0.14-0.63, p(trend) = 0.002, min p = 0.004, FDR p = 0.17). Our findings suggest that genetic polymorphisms in immune response genes might play a role in childhood leukemia development with limited biologic evidence.


Asunto(s)
Proteína de Dominio de Muerte Asociada a Fas/genética , Leucemia/genética , Leucemia/inmunología , Canales Catiónicos TRPM/genética , Adolescente , Niño , Preescolar , Proteína de Dominio de Muerte Asociada a Fas/inmunología , Femenino , Predisposición Genética a la Enfermedad , Genotipo , Humanos , Inmunidad/genética , Lactante , Recién Nacido , Corea (Geográfico) , Masculino , Polimorfismo de Nucleótido Simple , Riesgo , Canales Catiónicos TRPM/inmunología
17.
Immunol Res ; 51(2-3): 227-36, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22038529

RESUMEN

RIP1 is an adaptor serine/threonine kinase associated with the signaling complex of death receptors (DRs) including Fas, TNFR1, and TRAIL-Rs which can initiate apoptosis. While DRs are dispensable throughout development, RIP1 deletion results in perinatal lethality. The developmental defect caused by absence of RIP1 remains unexplained. In previous studies, RIP1-deficient hematopoietic progenitors failed to reconstitute the T cell compartment and our recent data indicate a new role for RIP1 in TCR-induced activation of the pro-survival NF-κB pathway. Here, we show that RIP1 is also critical for B cell development. In addition, RIP1(-/-) B cells stimulated through LPS/TLR4 are impaired in NF-κB activation but have no major defect in the Akt pathway. Recently, RIP1 has also emerged as a critical player in necrosis-like death, necroptosis, in various cell lines. We have demonstrated that RIP1 deficiency can reverse the embryonic and T cell proliferation defects in mice lacking FADD, a caspase adaptor protein, which indicates a potential role for RIP1 in mediating in vivo necroptosis. We provide an overview and discussion of the accumulating data revealing insights into the diverse functions of RIP1 in survival and death signaling in lymphocytes.


Asunto(s)
Proteína de Dominio de Muerte Asociada a Fas/inmunología , Proteínas Activadoras de GTPasa/inmunología , Linfocitos/inmunología , Necrosis/inmunología , Animales , Supervivencia Celular/genética , Proteína de Dominio de Muerte Asociada a Fas/genética , Proteínas Activadoras de GTPasa/genética , Humanos , Ratones , Ratones Noqueados , FN-kappa B/inmunología , Necrosis/genética , Proteínas Proto-Oncogénicas c-akt/inmunología , Transducción de Señal/inmunología
18.
Immunity ; 35(4): 572-82, 2011 Oct 28.
Artículo en Inglés | MEDLINE | ID: mdl-22000287

RESUMEN

Epidermal keratinocytes provide an essential structural and immunological barrier forming the first line of defense against potentially pathogenic microorganisms. Mechanisms regulating barrier integrity and innate immune responses in the epidermis are important for the maintenance of skin immune homeostasis and the pathogenesis of inflammatory skin diseases. Here, we show that epidermal keratinocyte-restricted deficiency of the adaptor protein FADD (FADD(E-KO)) induced severe inflammatory skin lesions in mice. The development of skin inflammation in FADD(E-KO) mice was triggered by RIP kinase 3 (RIP3)-mediated programmed necrosis (termed necroptosis) of FADD-deficient keratinocytes, which was partly dependent on the deubiquitinating enzyme CYLD and tumor necrosis factor (TNF)-TNF receptor 1 signaling. Collectively, our findings provide an in vivo experimental paradigm that regulation of necroptosis in keratinocytes is important for the maintenance of immune homeostasis and the prevention of chronic inflammation in the skin.


Asunto(s)
Apoptosis , Dermatitis/inmunología , Dermatitis/patología , Epidermis/inmunología , Proteína de Dominio de Muerte Asociada a Fas/inmunología , Queratinocitos/inmunología , Animales , Células Cultivadas , Dermatitis/metabolismo , Proteína de Dominio de Muerte Asociada a Fas/deficiencia , Homeostasis , Ratones , Ratones Noqueados , Factor 88 de Diferenciación Mieloide/metabolismo , Necrosis , Transducción de Señal
19.
Lupus ; 20(1): 71-9, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21078760

RESUMEN

Dysregulated neutrophil apoptosis may result in the development of autoimmune disease by contributing to nuclear autoantigen exposure, leading to autoantibody generation and a breakdown in immune tolerance. It has previously been shown that neutrophil apoptosis is increased in juvenile-onset systemic lupus erythematosus (JSLE). This study aims to investigate the pathways involved in JSLE serum-induced apoptosis. Caspases 3, 7-9, IAP1/2, XIAP and FADD mRNA levels and TRAIL R2, BID/tBID, caspase 8 and 9 protein expression were measured in neutrophils from JSLE patients (n = 14) and controls (n = 10). The mRNA levels of caspases 7-9 were significantly higher in JSLE neutrophils than in controls, whereas the mRNA levels of IAP1, IAP2 and XIAP were decreased (p < 0.05). A decrease in neutrophil apoptosis induced by JSLE serum was observed in the presence of caspase 8 and 9 inhibitors (p < 0.05), and the activity of caspases 8 and 9 increased over time. tBID protein expression increased following incubation with JSLE serum. These data focus specifically on the expression and activity of the main caspases in the intrinsic and extrinsic apoptotic pathways. Increased expression of factors involved in the downstream signalling of the extrinsic apoptotic pathway indicates a prominent involvement of this pathway in JSLE serum-induced apoptosis.


Asunto(s)
Apoptosis/inmunología , Lupus Eritematoso Sistémico/inmunología , Adolescente , Proteína Proapoptótica que Interacciona Mediante Dominios BH3/genética , Proteína Proapoptótica que Interacciona Mediante Dominios BH3/inmunología , Inhibidores de Caspasas , Caspasas/genética , Caspasas/inmunología , Niño , Proteína de Dominio de Muerte Asociada a Fas/genética , Proteína de Dominio de Muerte Asociada a Fas/inmunología , Femenino , Humanos , Proteínas Inhibidoras de la Apoptosis/genética , Proteínas Inhibidoras de la Apoptosis/inmunología , Masculino , Neutrófilos/citología , Neutrófilos/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptores del Ligando Inductor de Apoptosis Relacionado con TNF/genética , Receptores del Ligando Inductor de Apoptosis Relacionado con TNF/inmunología , Proteína Inhibidora de la Apoptosis Ligada a X/genética , Proteína Inhibidora de la Apoptosis Ligada a X/inmunología
20.
J Immunol ; 185(12): 7646-53, 2010 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-21068410

RESUMEN

Apoptosis of oligodendrocytes (ODCs), the myelin-producing glial cells in the CNS, plays a central role in demyelinating diseases such as multiple sclerosis and experimental autoimmune encephalomyelitis (EAE), an animal model of multiple sclerosis. To investigate the mechanism behind ODC apoptosis in EAE, we made use of conditional knockout mice lacking the adaptor protein FADD specifically in ODCs (FADD(ODC-KO)). FADD mediates apoptosis by coupling death receptors with downstream caspase activation. In line with this, ODCs from FADD(ODC-KO) mice were completely resistant to death receptor-induced apoptosis in vitro. In the EAE model, FADD(ODC-KO) mice followed an ameliorated clinical disease course in comparison with control littermates. Lymphocyte and macrophage infiltration into the spinal cord parenchyma was significantly reduced, as was the extent of demyelination and proinflammatory gene expression. Collectively, our data show that FADD is critical for ODC apoptosis and the development of autoimmune demyelinating disease.


Asunto(s)
Apoptosis/inmunología , Encefalomielitis Autoinmune Experimental/inmunología , Proteína de Dominio de Muerte Asociada a Fas/inmunología , Eliminación de Gen , Esclerosis Múltiple/inmunología , Oligodendroglía/inmunología , Animales , Apoptosis/genética , Encefalomielitis Autoinmune Experimental/genética , Encefalomielitis Autoinmune Experimental/metabolismo , Encefalomielitis Autoinmune Experimental/patología , Proteína de Dominio de Muerte Asociada a Fas/biosíntesis , Proteína de Dominio de Muerte Asociada a Fas/genética , Regulación de la Expresión Génica/genética , Regulación de la Expresión Génica/inmunología , Inflamación/genética , Inflamación/inmunología , Inflamación/metabolismo , Inflamación/patología , Linfocitos/inmunología , Linfocitos/metabolismo , Linfocitos/patología , Macrófagos/inmunología , Macrófagos/metabolismo , Macrófagos/patología , Ratones , Ratones Noqueados , Esclerosis Múltiple/genética , Esclerosis Múltiple/metabolismo , Esclerosis Múltiple/patología , Vaina de Mielina/inmunología , Vaina de Mielina/metabolismo , Vaina de Mielina/patología , Oligodendroglía/metabolismo , Oligodendroglía/patología , Médula Espinal/inmunología , Médula Espinal/metabolismo , Médula Espinal/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...