Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 61
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Nat Immunol ; 21(1): 54-64, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31819256

RESUMEN

Ptpn6 is a cytoplasmic phosphatase that functions to prevent autoimmune and interleukin-1 (IL-1) receptor-dependent, caspase-1-independent inflammatory disease. Conditional deletion of Ptpn6 in neutrophils (Ptpn6∆PMN) is sufficient to initiate IL-1 receptor-dependent cutaneous inflammatory disease, but the source of IL-1 and the mechanisms behind IL-1 release remain unclear. Here, we investigate the mechanisms controlling IL-1α/ß release from neutrophils by inhibiting caspase-8-dependent apoptosis and Ripk1-Ripk3-Mlkl-regulated necroptosis. Loss of Ripk1 accelerated disease onset, whereas combined deletion of caspase-8 and either Ripk3 or Mlkl strongly protected Ptpn6∆PMN mice. Ptpn6∆PMN neutrophils displayed increased p38 mitogen-activated protein kinase-dependent Ripk1-independent IL-1 and tumor necrosis factor production, and were prone to cell death. Together, these data emphasize dual functions for Ptpn6 in the negative regulation of p38 mitogen-activated protein kinase activation to control tumor necrosis factor and IL-1α/ß expression, and in maintaining Ripk1 function to prevent caspase-8- and Ripk3-Mlkl-dependent cell death and concomitant IL-1α/ß release.


Asunto(s)
Apoptosis/inmunología , Caspasa 8/inmunología , Neutrófilos/inmunología , Proteínas Quinasas/inmunología , Proteína Tirosina Fosfatasa no Receptora Tipo 6/metabolismo , Proteína Serina-Treonina Quinasas de Interacción con Receptores/inmunología , Animales , Caspasa 8/genética , Células Cultivadas , Eliminación de Gen , Inflamación/inmunología , Interleucina-1/inmunología , Interleucina-1alfa/metabolismo , Interleucina-1beta/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteína Tirosina Fosfatasa no Receptora Tipo 6/genética , Receptores Tipo I de Interleucina-1/inmunología , Factor de Necrosis Tumoral alfa/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
2.
Immunity ; 48(3): 570-583.e8, 2018 03 20.
Artículo en Inglés | MEDLINE | ID: mdl-29562203

RESUMEN

Polymorphisms in NFKB1 that diminish its expression have been linked to human inflammatory diseases and increased risk for epithelial cancers. The underlying mechanisms are unknown, and the link is perplexing given that NF-κB signaling reportedly typically exerts pro-tumorigenic activity. Here we have shown that NF-κB1 deficiency, even loss of a single allele, resulted in spontaneous invasive gastric cancer (GC) in mice that mirrored the histopathological progression of human intestinal-type gastric adenocarcinoma. Bone marrow chimeras revealed that NF-κB1 exerted tumor suppressive functions in both epithelial and hematopoietic cells. RNA-seq analysis showed that NF-κB1 deficiency resulted in aberrant JAK-STAT signaling, which dysregulated expression of effectors of inflammation, antigen presentation, and immune checkpoints. Concomitant loss of STAT1 prevented these immune abnormalities and GC development. These findings provide mechanistic insight into how polymorphisms that attenuate NFKB1 expression predispose humans to epithelial cancers, highlighting the pro-tumorigenic activity of STAT1 and identifying targetable vulnerabilities in GC.


Asunto(s)
Regulación Neoplásica de la Expresión Génica , Inflamación/genética , Inflamación/metabolismo , FN-kappa B/deficiencia , Factor de Transcripción STAT1/metabolismo , Neoplasias Gástricas/genética , Neoplasias Gástricas/metabolismo , Animales , Presentación de Antígeno/inmunología , Células de la Médula Ósea/metabolismo , Células de la Médula Ósea/patología , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/metabolismo , Modelos Animales de Enfermedad , Células Epiteliales/metabolismo , Células Epiteliales/patología , Redes Reguladoras de Genes , Humanos , Inflamación/patología , Ratones , Ratones Noqueados , Factor de Transcripción STAT1/deficiencia , Neoplasias Gástricas/inmunología , Neoplasias Gástricas/patología
3.
Genes Dev ; 32(21-22): 1420-1429, 2018 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-30366906

RESUMEN

Mutations in Trp53, prevalent in human cancer, are reported to drive tumorigenesis through dominant-negative effects (DNEs) over wild-type TRP53 function as well as neomorphic gain-of-function (GOF) activity. We show that five TRP53 mutants do not accelerate lymphomagenesis on a TRP53-deficient background but strongly synergize with c-MYC overexpression in a manner that distinguishes the hot spot Trp53 mutations. RNA sequencing revealed that the mutant TRP53 DNE does not globally repress wild-type TRP53 function but disproportionately impacts a subset of wild-type TRP53 target genes. Accordingly, TRP53 mutant proteins impair pathways for DNA repair, proliferation, and metabolism in premalignant cells. This reveals that, in our studies of lymphomagenesis, mutant TRP53 drives tumorigenesis primarily through the DNE, which modulates wild-type TRP53 function in a manner advantageous for neoplastic transformation.


Asunto(s)
Carcinogénesis/genética , Mutación , Proteína p53 Supresora de Tumor/genética , Animales , Linfoma/genética , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteína p53 Supresora de Tumor/metabolismo
4.
Immunity ; 45(3): 513-526, 2016 09 20.
Artículo en Inglés | MEDLINE | ID: mdl-27523270

RESUMEN

The kinases RIPK1 and RIPK3 and the pseudo-kinase MLKL have been identified as key regulators of the necroptotic cell death pathway, although a role for MLKL within the whole animal has not yet been established. Here, we have shown that MLKL deficiency rescued the embryonic lethality caused by loss of Caspase-8 or FADD. Casp8(-/-)Mlkl(-/-) and Fadd(-/-)Mlkl(-/-) mice were viable and fertile but rapidly developed severe lymphadenopathy, systemic autoimmune disease, and thrombocytopenia. These morbidities occurred more rapidly and with increased severity in Casp8(-/-)Mlkl(-/-) and Fadd(-/-)Mlkl(-/-) mice compared to Casp8(-/-)Ripk3(-/-) or Fadd(-/-)Ripk3(-/-) mice, respectively. These results demonstrate that MLKL is an essential effector of aberrant necroptosis in embryos caused by loss of Caspase-8 or FADD. Furthermore, they suggest that RIPK3 and/or MLKL may exert functions independently of necroptosis. It appears that non-necroptotic functions of RIPK3 contribute to the lymphadenopathy, autoimmunity, and excess cytokine production that occur when FADD or Caspase-8-mediated apoptosis is abrogated.


Asunto(s)
Apoptosis/fisiología , Enfermedades Autoinmunes/metabolismo , Muerte Celular/fisiología , Proteína de Dominio de Muerte Asociada a Fas/metabolismo , Proteínas Quinasas/metabolismo , Proteína Serina-Treonina Quinasas de Interacción con Receptores/metabolismo , Animales , Caspasa 8/metabolismo , Ratones , Ratones Endogámicos C57BL , Necrosis/metabolismo
5.
Immunity ; 42(5): 890-902, 2015 May 19.
Artículo en Inglés | MEDLINE | ID: mdl-25979420

RESUMEN

The mechanistic links between genetic variation and autoantibody production in autoimmune disease remain obscure. Autoimmune lymphoproliferative syndrome (ALPS) is caused by inactivating mutations in FAS or FASL, with autoantibodies thought to arise through failure of FAS-mediated removal of self-reactive germinal center (GC) B cells. Here we show that FAS is in fact not required for this process. Instead, FAS inactivation led to accumulation of a population of unconventional GC B cells that underwent somatic hypermutation, survived despite losing antigen reactivity, and differentiated into a large population of plasma cells that included autoantibody-secreting clones. IgE(+) plasma cell numbers, in particular, increased after FAS inactivation and a major cohort of ALPS-affected patients were found to have hyper-IgE. We propose that these previously unidentified cells, designated "rogue GC B cells," are a major driver of autoantibody production and provide a mechanistic explanation for the linked production of IgE and autoantibodies in autoimmune disease.


Asunto(s)
Autoanticuerpos/inmunología , Linfocitos B/citología , Centro Germinal/citología , Centro Germinal/inmunología , Inmunoglobulina E/inmunología , Receptor fas/inmunología , Animales , Autoanticuerpos/biosíntesis , Linfocitos B/inmunología , Ensayo de Inmunoadsorción Enzimática , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Humanos , Inmunoglobulina E/biosíntesis , Ratones , Reacción en Cadena de la Polimerasa , Receptor fas/deficiencia , Receptor fas/metabolismo
6.
Immunity ; 41(4): 646-56, 2014 Oct 16.
Artículo en Inglés | MEDLINE | ID: mdl-25367576

RESUMEN

Natural killer (NK) cells have been reported to control adaptive immune responses that occur in lymphoid organs at the early stages of immune challenge. The physiological purpose of such regulatory activity remains unclear, because it generally does not confer a survival advantage. We found that NK cells specifically eliminated activated CD4(+) T cells in the salivary gland during chronic murine cytomegalovirus (MCMV) infection. This was dependent on TNF-related apoptosis inducing ligand (TRAIL) expression by NK cells. Although NK cell-mediated deletion of CD4(+) T cells prolonged the chronicity of infection, it also constrained viral-induced autoimmunity. In the absence of this activity, chronic infection was associated with a Sjogren's-like syndrome characterized by focal lymphocytic infiltration into the glands, production of autoantibodies, and reduced saliva and tear secretion. Thus, NK cells are an important homeostatic control that balances the efficacy of adaptive immune responses with the risk of developing autoimmunity.


Asunto(s)
Autoinmunidad/inmunología , Linfocitos T CD4-Positivos/inmunología , Infecciones por Herpesviridae/inmunología , Células Asesinas Naturales/inmunología , Muromegalovirus/inmunología , Ligando Inductor de Apoptosis Relacionado con TNF/inmunología , Traslado Adoptivo , Animales , Apoptosis/inmunología , Enfermedad Crónica , Citotoxicidad Inmunológica/inmunología , Células Asesinas Naturales/trasplante , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Subfamilia K de Receptores Similares a Lectina de Células NK/biosíntesis , Glándulas Salivales/inmunología , Glándulas Salivales/patología , Glándulas Salivales/virología
7.
Immunity ; 39(3): 573-83, 2013 Sep 19.
Artículo en Inglés | MEDLINE | ID: mdl-24012421

RESUMEN

Activation-induced cell death (AICD) plays a critical role in immune homeostasis and tolerance. In T-cell-dependent humoral responses, AICD of B cells is initiated by Fas ligand (FasL) on T cells, stimulating the Fas receptor on B cells. In contrast, T-cell-independent B cell responses involve innate-type B lymphocytes, such as marginal zone (MZ) B cells, and little is known about the mechanisms that control AICD during innate B cell responses to Toll-like receptor (TLR) activation. Here, we show that MZ B cells undergo AICD in response to TLR4 activation in vivo. The transmembrane activator, calcium modulator, and cyclophilin ligand interactor (TACI) receptor and TLR4 cooperate to upregulate expression of both FasL and Fas on MZ B cells and also to repress inhibitors of Fas-induced apoptosis signaling. These findings demonstrate an unappreciated role for TACI and its ligands in the regulation of AICD during T-cell-independent B cell responses.


Asunto(s)
Apoptosis , Proteína Ligando Fas/metabolismo , Receptor Toll-Like 4/metabolismo , Proteína Activadora Transmembrana y Interactiva del CAML/metabolismo , Receptor fas/metabolismo , Animales , Receptor del Factor Activador de Células B/biosíntesis , Linfocitos B/inmunología , Activación Enzimática , Proteína Ligando Fas/biosíntesis , Lipopolisacáridos , Activación de Linfocitos/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteína Activadora Transmembrana y Interactiva del CAML/genética
8.
Gastroenterology ; 159(4): 1444-1458.e15, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32569771

RESUMEN

BACKGROUND & AIMS: Activity of nuclear factor κB transcription factors and signaling via signal transducer and activator of transcription (STAT) are frequently altered in gastric cancer cells. Mice lacking NFKB1 (Nfkb1-/- mice) develop invasive gastric cancer, and their gastric tissues have increased levels of cytokines, such as interleukin (IL) 6, IL22, IL11, and tumor necrosis factor (TNF), as well as increased activation of STAT1. We investigated whether these cytokines were required for STAT1 activation in gastric tissues of mice and critical for gastric tumorigenesis. METHODS: We crossed Nfkb1-/- mice with Il6-/-, Il22-/-, Il11Rα-/-, and Tnf-/- mice. Stomach tissues from compound mutant mice were analyzed by histology, immunoblotting, and RNA sequencing. Lymphoid, myeloid, and epithelial cells were isolated from stomachs, and the levels of cytokines were determined by flow cytometric analysis. RESULTS: Nfkb1-/- mice developed gastritis, oxyntic atrophy, gastric dysplasia, and invasive tumors, whereas Nfkb1-/-Stat1-/- mice did not, even when followed for as long as 2 years. The levels of Il6, Il11, Il22, and Tnf messenger RNA were increased in the body and antrum of the stomachs from Nfkb1-/- mice, from 3-6 months of age. However, Nfkb1-/-Il6-/-, Nfkb1-/-Il22-/-, and Nfkb1-/-Il11Rα-/- mice still developed gastric tumors, although the absence of IL11 receptor (IL11R) significantly reduced development of invasive gastric tumors. Stomachs from Nfkb1-/-Tnf-/- mice exhibited significantly less gastritis and oxyntic atrophy and fewer tumors than Nfkb1-/- mice. This correlated with reduced activation of STAT1 and STAT3 and fewer numbers of T cells and B cells infiltrating the gastric body. Loss of STAT1 or TNF significantly reduced expression of PD-L1 on epithelial and myeloid (CD11b+) cells in the gastric mucosa of Nfkb1-/- mice-indeed, to the levels observed on the corresponding cells from wild-type mice. CONCLUSIONS: In studies of gastric tumor development in knockout mice, we found that loss of NFKB1 causes increased expression of TNF in the stomach and thereby drives activation of STAT1, resulting in an inflammatory immune response and the development of gastric cancer. IL11R appears to be required for the progression of gastric tumors to the invasive stage. These findings suggest that inhibitors of TNF, and possibly also inhibitors of IL11/IL11Rα, might be useful in the treatment of gastric cancer.


Asunto(s)
Gastritis/patología , Subunidad p50 de NF-kappa B/metabolismo , Factor de Transcripción STAT1/metabolismo , Neoplasias Gástricas/etiología , Neoplasias Gástricas/patología , Factor de Necrosis Tumoral alfa/metabolismo , Animales , Carcinogénesis , Gastritis/etiología , Gastritis/metabolismo , Interleucina-11/metabolismo , Interleucina-6/metabolismo , Ratones , Transducción de Señal , Neoplasias Gástricas/metabolismo
9.
Immunity ; 37(3): 451-62, 2012 Sep 21.
Artículo en Inglés | MEDLINE | ID: mdl-22960223

RESUMEN

Although the proapoptotic BH3-only protein, Bim, is required for deletion of autoreactive thymocytes, Bim-deficient mice do not succumb to extensive organ-specific autoimmune disease. To determine whether other BH3-only proteins safeguard tolerance in the absence of Bim, we screened mice lacking Bim as well as other BH3-only proteins. Most strains showed no additional defects; however, mice deficient for both Puma and Bim spontaneously developed autoimmunity in multiple organs, and their T cells could transfer organ-specific autoimmunity. Puma- and Bim-double-deficient mice had a striking accumulation of mature, single-positive thymocytes, suggesting an additional defect in thymic deletion was the basis for disease. Transgenic mouse models of thymocyte deletion by peripheral neoantigens confirmed that the loss of Bim and Puma allowed increased numbers of autoreactive thymocytes to escape deletion. Our data show that Puma cooperates with Bim to impose a thymic-deletion checkpoint to peripheral self-antigens and cement the notion that defects in apoptosis alone are sufficient to cause autoimmune disease.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/inmunología , Autoantígenos/inmunología , Autoinmunidad/inmunología , Proteínas de la Membrana/inmunología , Proteínas Proto-Oncogénicas/inmunología , Timocitos/inmunología , Proteínas Supresoras de Tumor/inmunología , Animales , Apoptosis/genética , Apoptosis/inmunología , Proteínas Reguladoras de la Apoptosis/genética , Autoinmunidad/genética , Proteína 11 Similar a Bcl2 , Diferenciación Celular/genética , Diferenciación Celular/inmunología , Femenino , Citometría de Flujo , Masculino , Proteínas de la Membrana/genética , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas c-bcl-2/genética , Proteínas Proto-Oncogénicas c-bcl-2/inmunología , Autotolerancia/genética , Autotolerancia/inmunología , Linfocitos T/inmunología , Linfocitos T/metabolismo , Timocitos/metabolismo , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/inmunología , Proteínas Supresoras de Tumor/genética
10.
Immunity ; 36(4): 646-57, 2012 Apr 20.
Artículo en Inglés | MEDLINE | ID: mdl-22483802

RESUMEN

The immune system must distinguish viable cells from cells damaged by physical and infective processes. The damaged cell-recognition molecule Clec9A is expressed on the surface of the mouse and human dendritic cell subsets specialized for the uptake and processing of material from dead cells. Clec9A recognizes a conserved component within nucleated and nonnucleated cells, exposed when cell membranes are damaged. We have identified this Clec9A ligand as a filamentous form of actin in association with particular actin-binding domains of cytoskeletal proteins. We have determined the crystal structure of the human CLEC9A C-type lectin domain and propose a functional dimeric structure with conserved tryptophans in the ligand recognition site. Mutation of these residues ablated CLEC9A binding to damaged cells and to the isolated ligand complexes. We propose that Clec9A provides targeted recruitment of the adaptive immune system during infection and can also be utilized to enhance immune responses generated by vaccines.


Asunto(s)
Citoesqueleto de Actina/metabolismo , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Lectinas Tipo C/metabolismo , Receptores Inmunológicos/metabolismo , Receptores Mitogénicos/metabolismo , Actinas/metabolismo , Inmunidad Adaptativa , Animales , Sitios de Unión , Línea Celular , Membrana Celular/metabolismo , Células Dendríticas/citología , Femenino , Humanos , Lectinas Tipo C/química , Lectinas Tipo C/genética , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Estructura Secundaria de Proteína , Receptores Inmunológicos/genética , Receptores Mitogénicos/química , Receptores Mitogénicos/genética , Espectrina/metabolismo
11.
Immunol Cell Biol ; 97(8): 740-752, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31087793

RESUMEN

FOXP3+ regulatory T (Treg) cells are essential for immunological tolerance and immune homeostasis. Despite a great deal of interest in modulating their number and function for the treatment of autoimmune disease or cancer, the precise mechanisms that control the homeostasis of Treg cells remain unclear. We report a new ENU-induced mutant mouse, lack of costimulation (loco), with atopic dermatitis and Treg cell deficiency typical of Card11 loss-of-function mutants. Three distinct single nucleotide variants were found in the Card11 introns 2, 10 and 20 that cause the loss of CARD11 expression in these mutant mice. These mutations caused the loss of thymic-derived, Neuropilin-1+ (NRP1+ ) Treg cells in neonatal and adult loco mice; however, residual peripherally induced NRP1- Treg cells remained. These peripherally generated Treg cells could be expanded in vivo by the administration of IL-2:anti-IL-2 complexes, indicating that this key homeostatic signaling axis remained intact in CARD11-deficient Treg cells. Furthermore, these expanded Treg cells could mediate near-normal suppression of activated, conventional CD4+ T cells, suggesting that CARD11 is dispensable for Treg cell function. In addition to shedding light on the requirements for CARD11 in Treg cell homeostasis and function, these data reveal novel noncoding Card11 loss-of-function mutations that impair the expression of this critical immune-regulatory protein.


Asunto(s)
Proteínas Adaptadoras de Señalización CARD/deficiencia , Dermatitis Atópica/inmunología , Homeostasis/inmunología , Transducción de Señal/inmunología , Linfocitos T Reguladores/inmunología , Animales , Proteínas Adaptadoras de Señalización CARD/genética , Proteínas Adaptadoras de Señalización CARD/inmunología , Dermatitis Atópica/genética , Modelos Animales de Enfermedad , Etilnitrosourea/toxicidad , Factores de Transcripción Forkhead/inmunología , Factores de Transcripción Forkhead/metabolismo , Homeostasis/genética , Humanos , Intrones/efectos de los fármacos , Intrones/genética , Intrones/inmunología , Mutación con Pérdida de Función/efectos de los fármacos , Mutación con Pérdida de Función/inmunología , Ratones , Ratones Transgénicos , Mutagénesis/inmunología , Mutágenos/toxicidad , Neuropilina-1/inmunología , Neuropilina-1/metabolismo , Polimorfismo de Nucleótido Simple/efectos de los fármacos , Polimorfismo de Nucleótido Simple/inmunología , Transducción de Señal/genética , Linfocitos T Reguladores/metabolismo
12.
Nature ; 501(7466): 247-51, 2013 Sep 12.
Artículo en Inglés | MEDLINE | ID: mdl-24025841

RESUMEN

Successful infection by enteric bacterial pathogens depends on the ability of the bacteria to colonize the gut, replicate in host tissues and disseminate to other hosts. Pathogens such as Salmonella, Shigella and enteropathogenic and enterohaemorrhagic (EPEC and EHEC, respectively) Escherichia coli use a type III secretion system (T3SS) to deliver virulence effector proteins into host cells during infection that promote colonization and interfere with antimicrobial host responses. Here we report that the T3SS effector NleB1 from EPEC binds to host cell death-domain-containing proteins and thereby inhibits death receptor signalling. Protein interaction studies identified FADD, TRADD and RIPK1 as binding partners of NleB1. NleB1 expressed ectopically or injected by the bacterial T3SS prevented Fas ligand or TNF-induced formation of the canonical death-inducing signalling complex (DISC) and proteolytic activation of caspase-8, an essential step in death-receptor-induced apoptosis. This inhibition depended on the N-acetylglucosamine transferase activity of NleB1, which specifically modified Arg 117 in the death domain of FADD. The importance of the death receptor apoptotic pathway to host defence was demonstrated using mice deficient in the FAS signalling pathway, which showed delayed clearance of the EPEC-like mouse pathogen Citrobacter rodentium and reversion to virulence of an nleB mutant. The activity of NleB suggests that EPEC and other attaching and effacing pathogens antagonize death-receptor-induced apoptosis of infected cells, thereby blocking a major antimicrobial host response.


Asunto(s)
Escherichia coli Enteropatógena/metabolismo , Infecciones por Escherichia coli/metabolismo , Infecciones por Escherichia coli/microbiología , Proteínas de Escherichia coli/metabolismo , Tracto Gastrointestinal/microbiología , Transducción de Señal , Factores de Virulencia/metabolismo , Animales , Caspasa 8/metabolismo , Muerte Celular , Citrobacter rodentium/patogenicidad , Citrobacter rodentium/fisiología , Escherichia coli Enteropatógena/patogenicidad , Activación Enzimática , Infecciones por Escherichia coli/patología , Proteína Ligando Fas/antagonistas & inhibidores , Proteína Ligando Fas/metabolismo , Proteína de Dominio de Muerte Asociada a Fas/química , Proteína de Dominio de Muerte Asociada a Fas/metabolismo , Femenino , Células HEK293 , Células HeLa , Humanos , Masculino , Ratones , N-Acetilglucosaminiltransferasas/metabolismo , Estructura Terciaria de Proteína , Proteína Serina-Treonina Quinasas de Interacción con Receptores/química , Proteína Serina-Treonina Quinasas de Interacción con Receptores/metabolismo , Proteína de Dominio de Muerte Asociada a Receptor de TNF/química , Proteína de Dominio de Muerte Asociada a Receptor de TNF/metabolismo , Receptor fas/deficiencia , Receptor fas/metabolismo
13.
Proc Natl Acad Sci U S A ; 112(31): 9698-703, 2015 Aug 04.
Artículo en Inglés | MEDLINE | ID: mdl-26195802

RESUMEN

Rheumatoid arthritis (RA) and ankylosing spondylitis (AS) are chronic inflammatory diseases that together affect 2-3% of the population. RA and AS predominantly involve joints, but heart disease is also a common feature in RA and AS patients. Here we have studied a new spontaneous mutation that causes severe polyarthritis in bone phenotype spontaneous mutation 1 (BPSM1) mice. In addition to joint destruction, mutant mice also develop aortic root aneurism and aorto-mitral valve disease that can be fatal depending on the genetic background. The cause of the disease is the spontaneous insertion of a retrotransposon into the 3' untranslated region (3'UTR) of the tumor necrosis factor (TNF), which triggers its strong overexpression in myeloid cells. We found that several members of a family of RNA-binding, CCCH-containing zinc-finger proteins control TNF expression through its 3'UTR, and we identified a previously unidentified regulatory element in the UTR. The disease in BPSM1 mice is independent of the adaptive immune system and does not appear to involve inflammatory cytokines other than TNF. To our knowledge, this is the first animal model showing both polyarthritis and heart disease as a direct result of TNF deregulation. These results emphasize the therapeutic potential of anti-TNF drugs for the treatment of heart valve disease and identify potential therapeutic targets to control TNF expression and inflammation.


Asunto(s)
Regiones no Traducidas 3'/genética , Artritis/genética , Enfermedades de las Válvulas Cardíacas/genética , Mutagénesis Insercional/genética , Mutación/genética , Retroelementos/genética , Factor de Necrosis Tumoral alfa/genética , Aneurisma/patología , Animales , Insuficiencia de la Válvula Aórtica/complicaciones , Insuficiencia de la Válvula Aórtica/diagnóstico por imagen , Insuficiencia de la Válvula Aórtica/patología , Artritis/sangre , Artritis/diagnóstico por imagen , Artritis/patología , Secuencia de Bases , Trasplante de Médula Ósea , Quimiocinas/sangre , Enfermedad Crónica , Modelos Animales de Enfermedad , Fibrosis , Enfermedades de las Válvulas Cardíacas/sangre , Enfermedades de las Válvulas Cardíacas/diagnóstico por imagen , Enfermedades de las Válvulas Cardíacas/patología , Humanos , Inflamación/patología , Articulaciones/patología , Ratones Mutantes , Válvula Mitral/patología , Datos de Secuencia Molecular , Estabilidad del ARN/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Elementos de Nucleótido Esparcido Corto/genética , Factor de Necrosis Tumoral alfa/metabolismo , Ultrasonografía , Microtomografía por Rayos X , Dedos de Zinc/genética
14.
Reproduction ; 151(5): 553-62, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-26917450

RESUMEN

Apoptosis plays a prominent role during ovarian development by eliminating large numbers of germ cells from the female germ line. However, the precise mechanisms and regulatory proteins involved in germ cell death are yet to be determined. In this study, we characterised the role of the pro-apoptotic BH3-only protein, BCL2-modifying factor (BMF), in germ cell apoptosis in embryonic and neonatal mouse ovaries. BMF protein was immunohistochemically localised to germ cells at embryonic days 15.5 (E15.5) and E17.5 and postnatal day 1 (PN1), coincident with entry into the meiotic prophase, but was undetectable at E13.5, and only present at low levels at PN3 and PN5. Consistent with this expression pattern, loss of BMF in female mice was associated with a decrease in apoptosis at E15.5 and E17.5. Furthermore, increased numbers of germ cells were found in ovaries from Bmf(-/-) mice compared with WT animals at E15.5 and PN1. However, germ cell numbers were comparable between Bmf(-/-) and WT ovaries at PN3, PN5 and PN10. Collectively, these data indicate that BMF mediates foetal oocyte loss and its action limits the maximal number of germ cells attained in the developing ovary, but does not influence the number of primordial follicles initially established in ovarian reserve.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/fisiología , Apoptosis , Embrión de Mamíferos/patología , Células Germinativas/patología , Oocitos/patología , Oogénesis/fisiología , Ovario/patología , Animales , Western Blotting , Células Cultivadas , Embrión de Mamíferos/metabolismo , Femenino , Técnica del Anticuerpo Fluorescente , Células Germinativas/metabolismo , Técnicas para Inmunoenzimas , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Oocitos/metabolismo , Ovario/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2
15.
EMBO Rep ; 15(9): 982-90, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24990442

RESUMEN

A current paradigm proposes that mitochondrial damage is a critical determinant of NLRP3 inflammasome activation. Here, we genetically assess whether mitochondrial signalling represents a unified mechanism to explain how NLRP3 is activated by divergent stimuli. Neither co-deletion of the essential executioners of mitochondrial apoptosis BAK and BAX, nor removal of the mitochondrial permeability transition pore component cyclophilin D, nor loss of the mitophagy regulator Parkin, nor deficiency in MAVS affects NLRP3 inflammasome function. In contrast, caspase-8, a caspase essential for death-receptor-mediated apoptosis, is required for efficient Toll-like-receptor-induced inflammasome priming and cytokine production. Collectively, these results demonstrate that mitochondrial apoptosis is not required for NLRP3 activation, and highlight an important non-apoptotic role for caspase-8 in regulating inflammasome activation and pro-inflammatory cytokine levels.


Asunto(s)
Proteínas Portadoras/biosíntesis , Caspasa 8/biosíntesis , Inflamasomas/metabolismo , Mitocondrias/metabolismo , Apoptosis/genética , Autofagia/genética , Células de la Médula Ósea/metabolismo , Células de la Médula Ósea/patología , Proteínas Portadoras/genética , Caspasa 8/genética , Células Cultivadas , Peptidil-Prolil Isomerasa F , Ciclofilinas/antagonistas & inhibidores , Ciclofilinas/genética , Humanos , Interleucina-1beta/biosíntesis , Mitocondrias/patología , Mitofagia/genética , Proteína con Dominio Pirina 3 de la Familia NLR , Receptores Toll-Like/metabolismo , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/metabolismo
16.
J Pathol ; 236(3): 326-36, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25727407

RESUMEN

NF-κB signalling is an important factor in the development of inflammation-associated cancers. Mouse models of Helicobacter-induced gastric cancer and colitis-associated colorectal cancer have demonstrated that classical NF-κB signalling is an important regulator of these processes. In the stomach, it has also been demonstrated that signalling involving specific NF-κB proteins, including NF-κB1/p50, NF-κB2/p52, and c-Rel, differentially regulate the development of gastric pre-neoplasia. To investigate the effect of NF-κB subunit loss on colitis-associated carcinogenesis, we administered azoxymethane followed by pulsed dextran sodium sulphate to C57BL/6, Nfkb1(-/-), Nfkb2(-/-), and c-Rel(-/-) mice. Animals lacking the c-Rel subunit were more susceptible to colitis-associated cancer than wild-type mice, developing 3.5 times more colonic polyps per animal than wild-type mice. Nfkb2(-/-) mice were resistant to colitis-associated cancer, developing fewer polyps per colon than wild-type mice (median 1 compared to 4). To investigate the mechanisms underlying these trends, azoxymethane and dextran sodium sulphate were administered separately to mice of each genotype. Nfkb2(-/-) mice developed fewer clinical signs of colitis and exhibited less severe colitis and an attenuated cytokine response compared with all other groups following DSS administration. Azoxymethane administration did not fully suppress colonic epithelial mitosis in c-Rel(-/-) mice and less colonic epithelial apoptosis was also observed in this genotype compared to wild-type counterparts. These observations demonstrate different functions of specific NF-κB subunits in this model of colitis-associated carcinogenesis. NF-κB2/p52 is necessary for the development of colitis, whilst c-Rel-mediated signalling regulates colonic epithelial cell turnover following DNA damage.


Asunto(s)
Adenoma/metabolismo , Colitis/complicaciones , Neoplasias del Colon/metabolismo , Subunidad p50 de NF-kappa B/metabolismo , Subunidad p52 de NF-kappa B/metabolismo , Proteínas Proto-Oncogénicas c-rel/metabolismo , Adenoma/inducido químicamente , Adenoma/etiología , Animales , Azoximetano/toxicidad , Transformación Celular Neoplásica/metabolismo , Colitis/inducido químicamente , Neoplasias del Colon/inducido químicamente , Neoplasias del Colon/etiología , Citocinas/metabolismo , Sulfato de Dextran/toxicidad , Modelos Animales de Enfermedad , Susceptibilidad a Enfermedades , Células Epiteliales/metabolismo , Femenino , Inflamación , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Transducción de Señal
18.
Proc Natl Acad Sci U S A ; 110(7): 2599-604, 2013 Feb 12.
Artículo en Inglés | MEDLINE | ID: mdl-23349374

RESUMEN

Dysregulation of the "intrinsic" apoptotic pathway is associated with the development of cancer and autoimmune disease. Bak and Bax are two proapoptotic members of the Bcl-2 protein family with overlapping, essential roles in the intrinsic apoptotic pathway. Their activity is critical for the control of cell survival during lymphocyte development and homeostasis, best demonstrated by defects in thymic T-cell differentiation and peripheral lymphoid homeostasis caused by their combined loss. Because most bak(-/-)bax(-/-) mice die perinatally, the roles of Bax and Bak in immunological tolerance and prevention of autoimmune disease remain unclear. We show that mice reconstituted with a Bak/Bax doubly deficient hematopoietic compartment develop a fatal systemic lupus erythematosus-like autoimmune disease characterized by hypergammaglobulinemia, autoantibodies, lymphadenopathy, glomerulonephritis, and vasculitis. Importantly, these mice also develop a multiorgan autoimmune disease with autoantibodies against most solid glandular structures and evidence of glandular atrophy and necrotizing vasculitis. Interestingly, similar albeit less severe pathology was observed in mice containing a hematopoietic compartment deficient for only Bak, a phenotype reminiscent of the disease seen in patients with point mutations in BAK. These studies demonstrate a critical role for Bak and an ancillary role for Bax in safeguarding immunological tolerance and prevention of autoimmune disease. This suggests that direct activators of the intrinsic apoptotic pathway, such as BH3 mimetics, may be useful for treatment of diverse autoimmune diseases.


Asunto(s)
Apoptosis/inmunología , Enfermedades Autoinmunes/inmunología , Proteína Destructora del Antagonista Homólogo bcl-2/inmunología , Proteína X Asociada a bcl-2/inmunología , Animales , Autoanticuerpos/inmunología , Enfermedades Autoinmunes/patología , Western Blotting , Quimiocinas/sangre , Cruzamientos Genéticos , Citocinas/sangre , Ensayo de Inmunoadsorción Enzimática , Citometría de Flujo , Técnicas Histológicas , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microscopía Fluorescente , Proteína Destructora del Antagonista Homólogo bcl-2/deficiencia , Proteína X Asociada a bcl-2/deficiencia
19.
Nature ; 461(7264): 659-63, 2009 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-19794494

RESUMEN

Fas ligand (FasL), an apoptosis-inducing member of the TNF cytokine family, and its receptor Fas are critical for the shutdown of chronic immune responses and prevention of autoimmunity. Accordingly, mutations in their genes cause severe lymphadenopathy and autoimmune disease in mice and humans. FasL function is regulated by deposition in the plasma membrane and metalloprotease-mediated shedding. Here we generated gene-targeted mice that selectively lack either secreted FasL (sFasL) or membrane-bound FasL (mFasL) to resolve which of these forms is required for cell killing and to explore their hypothesized non-apoptotic activities. Mice lacking sFasL (FasL(Deltas/Deltas)) appeared normal and their T cells readily killed target cells, whereas T cells lacking mFasL (FasL(Deltam/Deltam)) could not kill cells through Fas activation. FasL(Deltam/Deltam) mice developed lymphadenopathy and hyper-gammaglobulinaemia, similar to FasL(gld/gld) mice, which express a mutant form of FasL that cannot bind Fas, but surprisingly, FasL(Deltam/Deltam) mice (on a C57BL/6 background) succumbed to systemic lupus erythematosus (SLE)-like autoimmune kidney destruction and histiocytic sarcoma, diseases that occur only rarely and much later in FasL(gld/gld) mice. These results demonstrate that mFasL is essential for cytotoxic activity and constitutes the guardian against lymphadenopathy, autoimmunity and cancer, whereas excess sFasL appears to promote autoimmunity and tumorigenesis through non-apoptotic activities.


Asunto(s)
Apoptosis , Membrana Celular/metabolismo , Proteína Ligando Fas/metabolismo , Receptor fas/metabolismo , Animales , Anticuerpos Antinucleares/inmunología , Citidina Desaminasa/metabolismo , Citotoxicidad Inmunológica , Proteína Ligando Fas/deficiencia , Proteína Ligando Fas/genética , Glomerulonefritis/metabolismo , Sarcoma Histiocítico/metabolismo , Hipergammaglobulinemia/metabolismo , Lupus Eritematoso Sistémico/metabolismo , Enfermedades Linfáticas/metabolismo , Ratones , Ratones Endogámicos C57BL , Mutación , Esplenomegalia/metabolismo , Linfocitos T/inmunología , Linfocitos T/metabolismo
20.
Proc Natl Acad Sci U S A ; 108(32): 13135-40, 2011 Aug 09.
Artículo en Inglés | MEDLINE | ID: mdl-21768356

RESUMEN

During immune responses, neutrophils must integrate survival and death signals from multiple sources to regulate their lifespan. Signals that activate either the Bcl-2- or death receptor-regulated apoptosis pathways can provide powerful stimuli for neutrophils to undergo cell death, but whether they act cooperatively in parallel or directly cross-talk in neutrophils is not known. Previous studies suggested that Bcl-2 family proteins are not required for Fas-induced cell death in neutrophils, but did not examine whether they could modulate its rapid onset. By monitoring the rate of change in neutrophil viability associated with activation of the Fas-triggered death receptor pathway using real-time cell imaging, we show that the Bcl-2-related proteins Bid, Bax, and Bak accelerate neutrophil apoptosis but are not essential for cell death. Increased Bcl-2 or Mcl-1 expression prevents efficient induction of apoptosis by Fas stimulation indicating that the Bcl-2-regulated apoptosis pathway can directly interfere with Fas-triggered apoptosis. Fas has been shown to initiate NFκB activation and gene transcription in cell lines, however gene transcription is not altered in Fas-activated Bid(-/-) neutrophils, indicating that apoptosis occurs independently of gene transcription in neutrophils. The specification of kinetics of neutrophil apoptosis by Bid impacts on the magnitude of neutrophil IL-1ß production, implicating a functional role for the Bcl-2-regulated pathway in controlling neutrophil responses to FasL. These data demonstrate that the intrinsic apoptosis pathway directly controls the kinetics of Fas-triggered apoptosis in neutrophils.


Asunto(s)
Apoptosis , Proteína Proapoptótica que Interacciona Mediante Dominios BH3/metabolismo , Neutrófilos/citología , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Proteína Destructora del Antagonista Homólogo bcl-2/metabolismo , Proteína X Asociada a bcl-2/metabolismo , Receptor fas/metabolismo , Animales , Apoptosis/efectos de los fármacos , Apoptosis/genética , Compuestos de Bifenilo/farmacología , Supervivencia Celular/efectos de los fármacos , Proteína Ligando Fas/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Imagenología Tridimensional , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteína 1 de la Secuencia de Leucemia de Células Mieloides , Neutrófilos/efectos de los fármacos , Neutrófilos/metabolismo , Nitrofenoles/farmacología , Piperazinas/farmacología , Transducción de Señal/efectos de los fármacos , Sulfonamidas/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA