Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Nat Commun ; 12(1): 4546, 2021 07 27.
Artículo en Inglés | MEDLINE | ID: mdl-34315884

RESUMEN

The NLRP3 inflammasome mediates the production of proinflammatory cytokines and initiates inflammatory cell death. Although NLRP3 is essential for innate immunity, aberrant NLRP3 inflammasome activation contributes to a wide variety of inflammatory diseases. Understanding the pathways that control NLRP3 activation will help develop strategies to treat these diseases. Here we identify WNK1 as a negative regulator of the NLRP3 inflammasome. Macrophages deficient in WNK1 protein or kinase activity have increased NLRP3 activation and pyroptosis compared with control macrophages. Mice with conditional knockout of WNK1 in macrophages have increased IL-1ß production in response to NLRP3 stimulation compared with control mice. Mechanistically, WNK1 tempers NLRP3 activation by balancing intracellular Cl- and K+ concentrations during NLRP3 activation. Collectively, this work shows that the WNK1 pathway has a critical function in suppressing NLRP3 activation and suggests that pharmacological inhibition of this pathway to treat hypertension might have negative clinical implications.


Asunto(s)
Cloruros/metabolismo , Inflamasomas/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Piroptosis , Proteína Quinasa Deficiente en Lisina WNK 1/metabolismo , Animales , Caspasa 1/metabolismo , Femenino , Imidazoles/farmacología , Inmunidad Innata/efectos de los fármacos , Interleucina-1beta/metabolismo , L-Lactato Deshidrogenasa/metabolismo , Lipopolisacáridos/farmacología , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Masculino , Ratones Endogámicos C57BL , Modelos Biológicos , Neutrófilos/efectos de los fármacos , Neutrófilos/metabolismo , Potasio/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Piroptosis/efectos de los fármacos , Pirrolidinas/farmacología , Tamoxifeno/farmacología , Proteína Quinasa Deficiente en Lisina WNK 1/antagonistas & inhibidores
2.
Nat Commun ; 10(1): 1689, 2019 04 11.
Artículo en Inglés | MEDLINE | ID: mdl-30976076

RESUMEN

Gasdermin E (GSDME/DFNA5) cleavage by caspase-3 liberates the GSDME-N domain, which mediates pyroptosis by forming pores in the plasma membrane. Here we show that GSDME-N also permeabilizes the mitochondrial membrane, releasing cytochrome c and activating the apoptosome. Cytochrome c release and caspase-3 activation in response to intrinsic and extrinsic apoptotic stimuli are significantly reduced in GSDME-deficient cells comparing with wild type cells. GSDME deficiency also accelerates cell growth in culture and in a mouse model of melanoma. Phosphomimetic mutation of the highly conserved phosphorylatable Thr6 residue of GSDME, inhibits its pore-forming activity, thus uncovering a potential mechanism by which GSDME might be regulated. Like GSDME-N, inflammasome-generated gasdermin D-N (GSDMD-N), can also permeabilize the mitochondria linking inflammasome activation to downstream activation of the apoptosome. Collectively, our results point to a role of gasdermin proteins in targeting the mitochondria to promote cytochrome c release to augment the mitochondrial apoptotic pathway.


Asunto(s)
Inflamasomas/metabolismo , Melanoma Experimental/patología , Mitocondrias/fisiología , Piroptosis/fisiología , Receptores de Estrógenos/metabolismo , Neoplasias Cutáneas/patología , Animales , Caspasa 3/metabolismo , Citocromos c/metabolismo , Fibroblastos , Técnicas de Inactivación de Genes , Células HEK293 , Células HeLa , Humanos , Macrófagos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Membranas Mitocondriales/metabolismo , Mutación , Fosforilación/fisiología , Cultivo Primario de Células , Dominios Proteicos/genética , Receptores de Estrógenos/genética , Treonina/metabolismo
3.
Nat Commun ; 9(1): 3001, 2018 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-30069026

RESUMEN

The NLRP3 inflammasome responds to infection and tissue damage, and rapidly escalates the intensity of inflammation by activating interleukin (IL)-1ß, IL-18 and cell death by pyroptosis. How the NLRP3 inflammasome is negatively regulated is poorly understood. Here we show that NLRP3 inflammasome activation is suppressed by sumoylation. NLRP3 is sumoylated by the SUMO E3-ligase MAPL, and stimulation-dependent NLRP3 desumoylation by the SUMO-specific proteases SENP6 and SENP7 promotes NLRP3 activation. Defective NLRP3 sumoylation, either by NLRP3 mutation of SUMO acceptor lysines or depletion of MAPL, results in enhanced caspase-1 activation and IL-1ß release. Conversely, depletion of SENP7 suppresses NLRP3-dependent ASC oligomerisation, caspase-1 activation and IL-1ß release. These data indicate that sumoylation of NLRP3 restrains inflammasome activation, and identify SUMO proteases as potential drug targets for the treatment of inflammatory diseases.


Asunto(s)
Inflamasomas/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Proteínas Modificadoras Pequeñas Relacionadas con Ubiquitina/metabolismo , Secuencia de Aminoácidos , Animales , Endopeptidasas/metabolismo , Células HEK293 , Humanos , Interleucina-1beta/metabolismo , Lisina/genética , Ratones , Mutación/genética , Proteína con Dominio Pirina 3 de la Familia NLR/química , Unión Proteica , Sumoilación , Ubiquitina-Proteína Ligasas/metabolismo
5.
Nat Commun ; 8: 14128, 2017 01 03.
Artículo en Inglés | MEDLINE | ID: mdl-28045099

RESUMEN

Apoptosis is a genetically regulated cell suicide programme mediated by activation of the effector caspases 3, 6 and 7. If apoptotic cells are not scavenged, they progress to a lytic and inflammatory phase called secondary necrosis. The mechanism by which this occurs is unknown. Here we show that caspase-3 cleaves the GSDMD-related protein DFNA5 after Asp270 to generate a necrotic DFNA5-N fragment that targets the plasma membrane to induce secondary necrosis/pyroptosis. Cells that express DFNA5 progress to secondary necrosis, when stimulated with apoptotic triggers such as etoposide or vesicular stomatitis virus infection, but disassemble into small apoptotic bodies when DFNA5 is deleted. Our findings identify DFNA5 as a central molecule that regulates apoptotic cell disassembly and progression to secondary necrosis, and provide a molecular mechanism for secondary necrosis. Because DFNA5-induced secondary necrosis and GSDMD-induced pyroptosis are dependent on caspase activation, we propose that they are forms of programmed necrosis.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/genética , Apoptosis/genética , Caspasa 3/genética , Necrosis/genética , Piroptosis/genética , Receptores de Estrógenos/genética , Animales , Apoptosis/efectos de los fármacos , Proteínas Reguladoras de la Apoptosis/metabolismo , Caspasa 3/metabolismo , Membrana Celular/efectos de los fármacos , Membrana Celular/metabolismo , Membrana Celular/virología , Etopósido/farmacología , Regulación de la Expresión Génica , Células HEK293 , Células Hep G2 , Humanos , Péptidos y Proteínas de Señalización Intracelular , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Macrófagos/virología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Necrosis/inducido químicamente , Necrosis/metabolismo , Proteínas de Unión a Fosfato , Cultivo Primario de Células , Piroptosis/efectos de los fármacos , Receptores de Estrógenos/metabolismo , Virus de la Estomatitis Vesicular Indiana/crecimiento & desarrollo , Virus de la Estomatitis Vesicular Indiana/patogenicidad
6.
Nat Commun ; 6: 7515, 2015 Jun 24.
Artículo en Inglés | MEDLINE | ID: mdl-26104484

RESUMEN

TLR2 promotes NLRP3 inflammasome activation via an early MyD88-IRAK1-dependent pathway that provides a priming signal (signal 1) necessary for activation of the inflammasome by a second potassium-depleting signal (signal 2). Here we show that TLR3 binding to dsRNA promotes post-translational inflammasome activation through intermediate and late TRIF/RIPK1/FADD-dependent pathways. Both pathways require the scaffolding but not the catalytic function of caspase-8 or RIPK1. Only the late pathway requires kinase competent RIPK3 and MLKL function. Mechanistically, FADD/caspase-8 scaffolding function provides a post-translational signal 1 in the intermediate pathway, whereas in the late pathway it helps the oligomerization of RIPK3, which together with MLKL provides both signal 1 and 2 for inflammasome assembly. Cytoplasmic dsRNA activates NLRP3 independent of TRIF, RIPK1, RIPK3 or mitochondrial DRP1, but requires FADD/caspase-8 in wildtype macrophages to remove RIPK3 inhibition. Our study provides a comprehensive analysis of pathways that lead to NLRP3 inflammasome activation in response to dsRNA.


Asunto(s)
Proteínas Portadoras/metabolismo , Caspasa 8/metabolismo , Macrófagos/metabolismo , Proteínas Quinasas/metabolismo , ARN Bicatenario/metabolismo , Receptor Toll-Like 3/metabolismo , Proteínas Adaptadoras del Transporte Vesicular/genética , Proteínas Adaptadoras del Transporte Vesicular/metabolismo , Animales , Proteínas Portadoras/genética , Caspasa 8/genética , Dinaminas/genética , Dinaminas/metabolismo , Proteína de Dominio de Muerte Asociada a Fas/genética , Proteína de Dominio de Muerte Asociada a Fas/metabolismo , Ratones , Ratones Noqueados , Mitocondrias/metabolismo , Factor 88 de Diferenciación Mieloide/genética , Factor 88 de Diferenciación Mieloide/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR , Proteínas Quinasas/genética , Proteína Serina-Treonina Quinasas de Interacción con Receptores/genética , Proteína Serina-Treonina Quinasas de Interacción con Receptores/metabolismo
7.
Immunol Cell Biol ; 93(6): 591-9, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25601272

RESUMEN

Th2 cytokine IL-4 has been previously shown to suppress the production of proinflammatory cytokines in monocytes. However, the underlying molecular mechanism by which IL-4 signaling antagonizes proinflammatory responses is poorly characterized. In particular, whether IL-4 can modulate inflammasome signaling remains unknown. Here, we provide evidence that IL-4 suppresses NLRP3-dependent caspase-1 activation and the subsequent IL-1ß secretion but does not inhibit absent in melanoma 2 (AIM2)- or NLRC4 (NOD-like receptor family, CARD domain-containing 4)-dependent caspase-1 activation in THP-1 and mouse bone marrow-derived macrophages. Upon lipopolysaccharide (LPS) or LPS/ATP stimulation, IL-4 markedly inhibited the assembly of NLRP3 inflammasome, including NLRP3-dependent ASC (apoptosis-associated speck-like protein containing a caspase recruitment domain) oligomerization, NLRP3-ASC interaction and NLRP3 speck-like oligomeric structure formation. The negative regulation of NLRP3 inflammasome by IL-4 was not due to the impaired mRNA or protein production of NLRP3 and proinflammatory cytokines. Supporting this observation, IL-4 attenuated NLRP3 inflammasome activation even in reconstituted NLRP3-expressing macrophages in which NLRP3 expression is not transcriptionally regulated by TLR-NF-κB signaling. Furthermore, the IL-4-mediated suppression of NLRP3 inflammasome was independent of STAT6-dependent transcription and mitochondrial reactive oxygen species (ROS). Instead, IL-4 inhibited subcellular redistribution of NLRP3 into mitochondria and microtubule polymerization upon NLRP3-activating stimulation. Our results collectively suggest that IL-4 could suppress NLRP3 inflammasome activation in a transcription-independent manner, thus providing an endogenous regulatory machinery to prevent excessive inflammasome activation.


Asunto(s)
Proteínas Portadoras/metabolismo , Inflamasomas/metabolismo , Interleucina-4/metabolismo , Transducción de Señal , Animales , Caspasa 1/metabolismo , Citocinas/genética , Citocinas/metabolismo , Activación Enzimática/efectos de los fármacos , Humanos , Mediadores de Inflamación/metabolismo , Interleucina-4/farmacología , Espacio Intracelular , Lipopolisacáridos/inmunología , Lipopolisacáridos/farmacología , Macrófagos/efectos de los fármacos , Macrófagos/inmunología , Macrófagos/metabolismo , Ratones , Proteína con Dominio Pirina 3 de la Familia NLR , Unión Proteica , Transporte de Proteínas , Especies Reactivas de Oxígeno/metabolismo , Factor de Transcripción STAT6/metabolismo , Transducción de Señal/efectos de los fármacos , Transcripción Genética/efectos de los fármacos
8.
J Immunol ; 192(8): 3881-8, 2014 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-24623131

RESUMEN

Caspase-1 activation is a central event in innate immune responses to many pathogenic infections and tissue damage. The NLRP3 inflammasome, a multiprotein scaffolding complex that assembles in response to two distinct steps, priming and activation, is required for caspase-1 activation. However, the detailed mechanisms of these steps remain poorly characterized. To investigate the process of LPS-mediated NLRP3 inflammasome priming, we used constitutively present pro-IL-18 as the caspase-1-specific substrate to allow study of the early events. We analyzed human monocyte caspase-1 activity in response to LPS priming, followed by activation with ATP. Within minutes of endotoxin priming, the NLRP3 inflammasome is licensed for ATP-induced release of processed IL-18, apoptosis-associated speck-forming complex containing CARD, and active caspase-1, independent of new mRNA or protein synthesis. Moreover, extracellular signal-regulated kinase 1 (ERK1) phosphorylation is central to the priming process. ERK inhibition and small interfering RNA-mediated ERK1 knockdown profoundly impair priming. In addition, proteasome inhibition prevents ERK phosphorylation and blocks priming. Scavenging reactive oxygen species with diphenylene iodonium also blocks both priming and ERK phosphorylation. These findings suggest that ERK1-mediated posttranslational modifications license the NLRP3 inflammasome to respond to the second signal ATP by inducing posttranslational events that are independent of new production of pro-IL-1ß and NOD-like receptor components.


Asunto(s)
Inflamasomas , Lipopolisacáridos/inmunología , Sistema de Señalización de MAP Quinasas , Complejo de la Endopetidasa Proteasomal/metabolismo , Proteínas Portadoras/metabolismo , Caspasa 1/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Inmunidad Innata , Inflamación/genética , Inflamación/inmunología , Inflamación/metabolismo , Interleucina-18/metabolismo , Interleucina-1beta/metabolismo , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Proteína Quinasa 1 Activada por Mitógenos/genética , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/genética , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Modelos Biológicos , Monocitos/inmunología , Monocitos/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR , Oxidantes/farmacología , Inhibidores de Proteínas Quinasas/farmacología
9.
J Immunol ; 191(8): 4358-66, 2013 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-24048902

RESUMEN

NLRP3 assembles an inflammasome complex that activates caspase-1 upon sensing various danger signals derived from pathogenic infection, tissue damage, and environmental toxins. How NLRP3 senses these various stimuli is still poorly understood, but mitochondria and mitochondrial reactive oxygen species have been proposed to play a critical role in NLRP3 activation. In this article, we provide evidence that the mitochondrial antiviral signaling protein MAVS associates with NLRP3 and facilitates its oligomerization leading to caspase-1 activation. In reconstituted 293T cells, full-length MAVS promoted NLRP3-dependent caspase-1 activation, whereas a C-terminal transmembrane domain-truncated mutant of MAVS (MAVS-ΔTM) did not. MAVS, but not MAVS-ΔTM, interacted with NLRP3 and triggered the oligomerization of NLRP3, suggesting that mitochondrial localization of MAVS and intact MAVS signaling are essential for activating the NLRP3 inflammasome. Supporting this, activation of MAVS signaling by Sendai virus infection promoted NLRP3-dependent caspase-1 activation, whereas knocking down MAVS expression clearly attenuated the activation of NLRP3 inflammasome by Sendai virus in THP-1 and mouse macrophages. Taken together, our results suggest that MAVS facilitates the recruitment of NLRP3 to the mitochondria and may enhance its oligomerization and activation by bringing it in close proximity to mitochondrial reactive oxygen species.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas Portadoras/metabolismo , Macrófagos/inmunología , Mitocondrias/metabolismo , Infecciones por Respirovirus/inmunología , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Células de la Médula Ósea/inmunología , Células de la Médula Ósea/metabolismo , Caspasa 1/metabolismo , Línea Celular , Activación Enzimática , Células HEK293 , Humanos , Inflamasomas/inmunología , Macrófagos/metabolismo , Ratones , Proteína con Dominio Pirina 3 de la Familia NLR , Interferencia de ARN , ARN Interferente Pequeño , Especies Reactivas de Oxígeno/metabolismo , Virus Sendai/inmunología , Transducción de Señal/inmunología
10.
J Immunol ; 191(8): 3995-9, 2013 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-24043892

RESUMEN

Activation of the NLRP3 inflammasome by diverse stimuli requires a priming signal from TLRs and an activation signal from purinergic receptors or pore-forming toxins. In this study, we demonstrate, through detailed analysis of NLRP3 activation in macrophages deficient in key downstream TLR signaling molecules, that MyD88 is required for an immediate early phase, whereas Toll/IL-1R domain-containing adapter inducing IFN-ß is required for a subsequent intermediate phase of posttranslational NLRP3 activation. Both IL-1R-associated kinase (IRAK) 1 and IRAK4 are critical for rapid activation of NLRP3 through the MyD88 pathway, but only IRAK1 is partially required in the Toll/IL-1R domain-containing adapter inducing IFN-ß pathway. IRAK1 and IRAK4 are also required for rapid activation of NLRP3 by Listeria monocytogenes, as deletion of IRAK1 or IRAK4 led to defective inflammasome activation. These findings define the pathways that lead to rapid NLRP3 activation and identify IRAK1 as a critical mediator of a transcription-independent,inflammasome-dependent early warning response to pathogenic infection.


Asunto(s)
Proteínas Portadoras/metabolismo , Inflamasomas , Quinasas Asociadas a Receptores de Interleucina-1/inmunología , Quinasas Asociadas a Receptores de Interleucina-1/metabolismo , Factor 88 de Diferenciación Mieloide/metabolismo , Receptores Toll-Like/inmunología , Proteínas Adaptadoras del Transporte Vesicular/metabolismo , Animales , Activación Enzimática , Interferón beta/metabolismo , Listeria monocytogenes/inmunología , Listeria monocytogenes/metabolismo , Macrófagos/enzimología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteína con Dominio Pirina 3 de la Familia NLR , Receptores de Interleucina-1/metabolismo , Transducción de Señal , Receptores Toll-Like/metabolismo
11.
J Biol Chem ; 288(16): 11378-83, 2013 Apr 19.
Artículo en Inglés | MEDLINE | ID: mdl-23479736

RESUMEN

Human pyrin with gain-of-function mutations in its B30.2/SPRY domain causes the autoinflammatory disease familial Mediterranean fever by assembling an ASC-dependent inflammasome that activates caspase-1. Wild-type human pyrin can also form an inflammasome complex with ASC after engagement by autoinflammatory PSTPIP1 mutants. How the pyrin inflammasome is activated in the absence of disease-associated mutations is not yet known. We report here that ribotoxic stress triggers the assembly of the human pyrin inflammasome, leading to ASC oligomerization and caspase-1 activation in THP-1 macrophages and in a 293T cell line stably reconstituted with components of the pyrin inflammasome. Knockdown of pyrin and selective inhibition of p38 MAPK greatly attenuated caspase-1 activation by ribotoxic stress, whereas expression of the conditional mutant ΔMEKK3:ER* allowed the activation of caspase-1 without ribotoxic stress. Disruption of microtubules by colchicine also inhibited pyrin inflammasome activation by ribotoxic stress. Together, our results indicate that ribotoxic stress activates the human pyrin inflammasome through a mechanism that requires p38 MAPK signaling and microtubule stability.


Asunto(s)
Proteínas del Citoesqueleto/metabolismo , Inflamasomas/metabolismo , Sistema de Señalización de MAP Quinasas/fisiología , Estrés Fisiológico/fisiología , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Caspasa 1/genética , Caspasa 1/metabolismo , Línea Celular , Colchicina/farmacología , Proteínas del Citoesqueleto/genética , Humanos , Inflamasomas/genética , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Microtúbulos/genética , Microtúbulos/metabolismo , Mutación , Pirina , Estrés Fisiológico/efectos de los fármacos , Moduladores de Tubulina/farmacología , Proteínas Quinasas p38 Activadas por Mitógenos/genética
12.
Cancer Lett ; 331(2): 183-91, 2013 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-23321501

RESUMEN

Apoptosis-associated speck-like protein containing a caspase recruitment domain (ASC), an essential component of the inflammasome complex, is frequently silenced by epigenetic methylation in many tumor cells. Here, we demonstrate that restoration of ASC expression in human colorectal cancer DLD-1 cells, in which ASC is silenced by aberrant methylation, potentiated cell death mediated by DNA damaging agent. Contrarily, ASC knockdown in HT-29 cells rendered cells less susceptible to etoposide toxicity. The increased susceptibility of ASC-expressing DLD-1 cells to genotoxic stress was independent of inflammasome or caspase activation, but partially dependent on mitochondrial ROS production and JNK activation. Thus, our data suggest that ASC expression in cancer cells is an important factor in determining their susceptibility to chemotherapy.


Asunto(s)
Apoptosis/efectos de los fármacos , Caspasas/metabolismo , Neoplasias Colorrectales/patología , Proteínas del Citoesqueleto/metabolismo , Mutágenos/toxicidad , Secuencia de Bases , Proteínas Adaptadoras de Señalización CARD , Línea Celular Tumoral , Neoplasias Colorrectales/enzimología , Metilación de ADN , Cartilla de ADN , Etopósido/farmacología , Técnica del Anticuerpo Fluorescente , Células HT29 , Humanos , Inflamasomas/metabolismo , Sistema de Señalización de MAP Quinasas , Mitocondrias/efectos de los fármacos , Mitocondrias/enzimología , Mitocondrias/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
13.
Autophagy ; 9(3): 420-1, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23242108

RESUMEN

HTRA2/OMI is an ATP-independent serine protease located in the intermembrane space of the mitochondria and is thought to function as a protein quality control protease. Our previous studies showed that loss of the enzymatic activity of HTRA2 due to a Ser276Cys missense mutation in its catalytic domain is associated with early onset neurodegeneration, multiple tissue atrophy and premature lethality in homozygous htra2 (mnd2) mice, suggesting that HTRA2 is neuroprotective. To further investigate the role of HTRA2 in neuronal cell survival and the impact of its loss of function in non-neuronal tissues of adult mice, we generated transgenic htra2 (mnd2) mice expressing a neuron-targeted human HTRA2 transgene. Notably, this HTRA2 transgene rescues htra2 (mnd2) mice from early onset neurodegeneration, and other phenotypic abnormalities and prevents their early death, indicating that HTRA2 activity in neuronal mitochondria is important for neuronal cell survival. However, as the rescued htra2 (mnd2) mice grow older they exhibit specific phenotypic abnormalities indicative of premature aging. These include premature weight loss, osteoporosis, lordokyphosis, muscle atrophy, heart enlargement, increased autophagy and reduced life span. There is also a significant increase in the levels of clonally expanded mitochondrial DNA (mtDNA) deletions in their tissues. Our findings suggest that HTRA2-regulated protein quality control in the intermembrane space of mitochondria is important for the maintenance of mitochondrial homeostasis, and loss of HTRA2 activity can lead to both neurodegeneration and aging.


Asunto(s)
Envejecimiento , Mitocondrias/metabolismo , Proteínas Mitocondriales/metabolismo , Neuronas/metabolismo , Serina Endopeptidasas/metabolismo , Adenosina Trifosfato/metabolismo , Animales , Apoptosis , Supervivencia Celular , ADN Mitocondrial/genética , Serina Peptidasa A2 que Requiere Temperaturas Altas , Humanos , Ratones , Ratones Transgénicos , Proteínas Mitocondriales/genética , Enfermedades Neurodegenerativas/genética , ARN Mensajero/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Serina Endopeptidasas/genética
14.
J Biol Chem ; 287(43): 36617-22, 2012 Oct 19.
Artículo en Inglés | MEDLINE | ID: mdl-22948162

RESUMEN

The NLRP3 inflammasome is a key component of the innate immune response to pathogenic infection and tissue damage. It is also involved in the pathogenesis of a number of human diseases, including gouty arthritis, silicosis, atherosclerosis, and type 2 diabetes. The assembly of the NLRP3 inflammasome requires a priming signal derived from pattern recognition or cytokine receptors, followed by a second signal derived from extracellular ATP, pore-forming toxins, or crystalline materials. How these two signals activate the NLRP3 inflammasome is not yet clear. Here, we show that in mouse macrophages, signaling by the pattern recognition receptor TLR4 through MyD88 can rapidly and non-transcriptionally prime NLRP3 by stimulating its deubiquitination. This process is dependent on mitochondrial reactive oxygen species production and can be inhibited by antioxidants. We further show that signaling by ATP can also induce deubiquitination of NLRP3 by a mechanism that is not sensitive to antioxidants. Pharmacological inhibition of NLRP3 deubiquitination completely blocked NLRP3 activation in both mouse and human cells, indicating that deubiquitination of NLRP3 is required for its activation. Our findings suggest that NLRP3 is activated by a two-step deubiquitination mechanism initiated by Toll-like receptor signaling and mitochondrial reactive oxygen species and further potentiated by ATP, which could explain how NLRP3 is activated by diverse danger signals.


Asunto(s)
Proteínas Portadoras/metabolismo , Inflamasomas/metabolismo , Macrófagos/metabolismo , Transducción de Señal , Ubiquitinación , Adenosina Trifosfato/genética , Adenosina Trifosfato/metabolismo , Animales , Proteínas Portadoras/genética , Células Cultivadas , Humanos , Inflamasomas/genética , Inflamación/genética , Inflamación/metabolismo , Inflamación/patología , Macrófagos/patología , Ratones , Ratones Noqueados , Mitocondrias/genética , Mitocondrias/metabolismo , Factor 88 de Diferenciación Mieloide/genética , Factor 88 de Diferenciación Mieloide/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR , Especies Reactivas de Oxígeno/metabolismo , Receptor Toll-Like 4/genética , Receptor Toll-Like 4/metabolismo
15.
J Immunol ; 187(9): 4890-9, 2011 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-21957143

RESUMEN

Streptococcus pneumoniae is a Gram-positive, extracellular bacterium that is responsible for significant mortality and morbidity worldwide. Pneumolysin (PLY), a cytolysin produced by all clinical isolates of the pneumococcus, is one of the most important virulence factors of this pathogen. We have previously reported that PLY is an essential factor for activation of caspase-1 and consequent secretion of IL-1ß and IL-18 in macrophages infected with S. pneumoniae. However, the host molecular factors involved in caspase-1 activation are still unclear. To further elucidate the mechanism of caspase-1 activation in macrophages infected with S. pneumoniae, we examined the involvement of inflammasomes in inducing this cellular response. Our study revealed that apoptosis-associated specklike protein containing a caspase recruitment domain (ASC), an adaptor protein for inflammasome receptors such as nucleotide-binding oligomerization domain-like receptor family, pyrin domain containing 3 (NLRP3) and absent in melanoma 2 (AIM2), is essentially required for the induction of caspase-1 activation by S. pneumoniae. Caspase-1 activation was partially impaired in NLRP3(-/-) macrophages, whereas knockdown and knockout of AIM2 resulted in a clear decrease in caspase-1 activation in response to S. pneumoniae. These results suggest that ASC inflammasomes, including AIM2 and NLRP3, are critical for caspase-1 activation induced by S. pneumoniae. Furthermore, ASC(-/-) mice were more susceptible than wild-type mice to S. pneumoniae, with impaired secretion of IL-1ß and IL-18 into the bronchoalveolar lavage after intranasal infection, suggesting that ASC inflammasomes contribute to the protection of host from infection with PLY-producing S. pneumoniae.


Asunto(s)
Caspasa 1/metabolismo , Proteínas del Citoesqueleto/fisiología , Inmunidad Innata , Inflamasomas/fisiología , Infecciones Neumocócicas/inmunología , Infecciones Neumocócicas/metabolismo , Animales , Proteínas Reguladoras de la Apoptosis , Proteínas Bacterianas/antagonistas & inhibidores , Proteínas Bacterianas/biosíntesis , Proteínas Adaptadoras de Señalización CARD , Proteínas Portadoras/fisiología , Caspasa 1/deficiencia , Caspasa 1/genética , Línea Celular , Línea Celular Transformada , Células Cultivadas , Proteínas del Citoesqueleto/deficiencia , Proteínas del Citoesqueleto/genética , Proteínas de Unión al ADN , Resistencia a la Enfermedad/inmunología , Activación Enzimática/inmunología , Femenino , Interleucina-18/metabolismo , Interleucina-1beta/metabolismo , Ratones , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Ratones Noqueados , Proteína con Dominio Pirina 3 de la Familia NLR , Proteínas Nucleares/fisiología , Infecciones Neumocócicas/enzimología , Estreptolisinas/antagonistas & inhibidores , Estreptolisinas/biosíntesis
16.
J Immunol ; 185(5): 2670-4, 2010 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-20679532

RESUMEN

The mechanisms by which the intracellular pathogen Francisella tularensis evades innate immunity are not well defined. We have identified a gene with homology to Escherichia coli mviN, a putative lipid II flippase, which F. tularensis uses to evade activation of innate immune pathways. Infection of mice with a F. tularensis mviN mutant resulted in improved survival and decreased bacterial burdens compared to infection with wild-type F. tularensis. The mviN mutant also induced increased absent in melanoma 2 inflammasome-dependent IL-1beta secretion and cytotoxicity in macrophages. The compromised in vivo virulence of the mviN mutant depended upon inflammasome activation, as caspase 1- and apoptosis-associated speck-like protein containing a caspase recruitment domain-deficient mice did not exhibit preferential survival following infection. This study demonstrates that mviN limits F. tularensis-induced absent in melanoma 2 inflammasome activation, which is critical for its virulence in vivo.


Asunto(s)
Proteínas Bacterianas/genética , Regulación hacia Abajo/inmunología , Francisella tularensis/genética , Activación de Macrófagos/inmunología , Macrófagos/inmunología , Mutación , Proteínas Nucleares/metabolismo , Regulación hacia Arriba/inmunología , Factores de Virulencia/genética , Animales , Proteínas Bacterianas/fisiología , Vacunas Bacterianas/genética , Vacunas Bacterianas/inmunología , Células de la Médula Ósea/citología , Células de la Médula Ósea/inmunología , Proteínas de Unión al ADN , Regulación hacia Abajo/genética , Francisella tularensis/inmunología , Francisella tularensis/patogenicidad , Mediadores de Inflamación/metabolismo , Mediadores de Inflamación/fisiología , Activación de Macrófagos/genética , Macrófagos/microbiología , Macrófagos/patología , Melanoma/inmunología , Ratones , Ratones Noqueados , Mutación/genética , Mutación/inmunología , Regulación hacia Arriba/genética , Virulencia/genética , Virulencia/inmunología , Factores de Virulencia/fisiología
17.
J Clin Immunol ; 30(5): 693-702, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20490635

RESUMEN

Infection with Listeria monocytogenes can cause meningitis and septicemia in newborn, elderly, or immunocompromised individuals. Pregnant women are particularly susceptible to Listeria, leading to a potentially fatal infection. Cytosolic Listeria activates the proinflammatory caspase-1 and induces the processing and secretion of interleukins IL-1beta and IL-18 as well as caspase-1-dependent pyroptosis. This study elucidates the role of various inflammasome components of host macrophages in the proinflammatory response to infection with Listeria. Here, we have used macrophages from AIM2-, NLRC4-, NLRP3-, and ASC-deficient mice to demonstrate that AIM2, NLRC4, and NLRP3 inflammasomes as well as the adaptor protein ASC all contribute to activation of caspase-1 in Listeria-infected macrophages. We show that Listeria DNA, which escapes into the cytosol of infected macrophages, triggers AIM2 oligomerization, caspase-1 activation, and pyroptosis. Interestingly, we found that flagellin-deficient Listeria, like Francisella tularensis, is recognized primarily by the AIM2 inflammasome, as no caspase-1 activation or cell death was observed in AIM2-deficient macrophages infected with this Listeria mutant. We further show that prior priming of NLRC4-deficient macrophages with LPS is sufficient for Listeria-induced caspase-1 activation in these macrophages, suggesting that TLR4 signaling is important for activation of the AIM2 and NLRP3 inflammasomes by Listeria in the absence of NLRC4. Taken together, our results indicate that Listeria infection is sensed by multiple inflammasomes that collectively orchestrate a robust caspase-1 activation and proinflammatory response.


Asunto(s)
Inflamasomas/inmunología , Interleucina-1beta/metabolismo , Listeria monocytogenes/inmunología , Listeriosis/inmunología , Macrófagos/metabolismo , Animales , Proteínas Reguladoras de la Apoptosis/genética , Proteínas Reguladoras de la Apoptosis/metabolismo , Proteínas de Unión al Calcio/genética , Proteínas de Unión al Calcio/metabolismo , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Caspasa 1/metabolismo , Línea Celular Transformada , ADN Bacteriano/farmacología , Proteínas de Unión al ADN , Activación Enzimática/efectos de los fármacos , Activación Enzimática/genética , Flagelina/genética , Flagelina/inmunología , Interleucina-1beta/inmunología , Listeria monocytogenes/genética , Listeria monocytogenes/patogenicidad , Listeriosis/genética , Listeriosis/metabolismo , Macrófagos/inmunología , Macrófagos/patología , Ratones , Ratones Noqueados , Mutación/genética , Proteína con Dominio Pirina 3 de la Familia NLR , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Multimerización de Proteína/efectos de los fármacos , Multimerización de Proteína/genética
18.
Nat Immunol ; 11(5): 385-93, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20351693

RESUMEN

Francisella tularensis, the causative agent of tularemia, infects host macrophages, which triggers production of the proinflammatory cytokines interleukin 1beta (IL-1beta) and IL-18. We elucidate here how host macrophages recognize F. tularensis and elicit this proinflammatory response. Using mice deficient in the DNA-sensing inflammasome component AIM2, we demonstrate here that AIM2 is required for sensing F. tularensis. AIM2-deficient mice were extremely susceptible to F. tularensis infection, with greater mortality and bacterial burden than that of wild-type mice. Caspase-1 activation, IL-1beta secretion and cell death were absent in Aim2(-/-) macrophages in response to F. tularensis infection or the presence of cytoplasmic DNA. Our study identifies AIM2 as a crucial sensor of F. tularensis infection and provides genetic proof of its critical role in host innate immunity to intracellular pathogens.


Asunto(s)
Francisella tularensis/inmunología , Inmunidad Innata , Macrófagos/metabolismo , Complejos Multiproteicos/metabolismo , Proteínas Nucleares/inmunología , Proteínas Nucleares/metabolismo , Tularemia/inmunología , Animales , Señalización del Calcio/inmunología , Caspasa 1/genética , Caspasa 1/inmunología , Caspasa 1/metabolismo , Células Cultivadas , Proteínas de Unión al ADN , Francisella tularensis/patogenicidad , Humanos , Factor 3 Regulador del Interferón/genética , Factor 3 Regulador del Interferón/inmunología , Factor 3 Regulador del Interferón/metabolismo , Interferón Tipo I/inmunología , Interleucina-1beta/biosíntesis , Interleucina-1beta/genética , Interleucina-1beta/inmunología , L-Lactato Deshidrogenasa/genética , L-Lactato Deshidrogenasa/inmunología , L-Lactato Deshidrogenasa/metabolismo , Macrófagos/inmunología , Macrófagos/patología , Ratones , Ratones Noqueados , Complejos Multiproteicos/genética , Complejos Multiproteicos/inmunología , Proteínas Nucleares/genética , Multimerización de Proteína , Tularemia/genética , Tularemia/metabolismo
19.
Arthritis Rheum ; 62(4): 1176-85, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20131254

RESUMEN

OBJECTIVE: To gain insight into the pathophysiology of an atypical familial form of an autoinflammatory disorder, characterized by autosomal-dominant sensorineural hearing loss, systemic inflammation, increased secretion of interleukin-1beta (IL-1beta), and the absence of any cutaneous manifestations, and to assess the functional consequences of a missense mutation identified in the leucine-rich repeat (LRR) domain of NLRP3. METHODS: Microsatellite markers were used to test the familial segregation of the NLRP3 locus with the disease phenotype. All NLRP3 exons were screened for mutations by sequencing. Functional assays were performed in HEK 293T cells to determine the effects of mutated (versus normal) NLRP3 proteins on NF-kappaB activation, caspase 1 signaling, and speck formation. RESULTS: A heterozygous NLRP3 missense mutation (p.Tyr859Cys) was identified in exon 6, which encodes the LRR domain of the protein. This mutation was found to segregate with the disease phenotype within the family, and had a moderate activating effect on speck formation and procaspase 1 processing and did not alter the inhibitory properties of NLRP3 on NF-kappaB signaling. CONCLUSION: This report is the first to describe a familial form of a cryopyrinopathy associated with a mutation outside of exon 3 of NLRP3. This finding, together with the known efficacy of anti-IL-1 treatments in these disorders, underlines the importance of screening all exons of NLRP3 in patients who present with atypical manifestations. In addition, the gain of function associated with this mutation in terms of activation of caspase 1 signaling was consistent with the observed inflammatory phenotype. Therefore, this study of the functional consequences of an LRR mutation sheds new light on the clinical relevance of in vitro assays.


Asunto(s)
Proteínas Portadoras/genética , Mutación de Línea Germinal , Enfermedades Autoinflamatorias Hereditarias/genética , Secuencia de Aminoácidos , Línea Celular , Citocinas/sangre , ADN/genética , Cartilla de ADN , Femenino , Variación Genética , Enfermedades Autoinflamatorias Hereditarias/fisiopatología , Humanos , Interleucina-1beta/sangre , Riñón/embriología , Masculino , Monocitos/patología , Madres , Mutación Missense , Proteína con Dominio Pirina 3 de la Familia NLR , Plásmidos , Hermanos
20.
J Biol Chem ; 285(13): 9792-9802, 2010 Mar 26.
Artículo en Inglés | MEDLINE | ID: mdl-20093358

RESUMEN

Activation of the inflammasome generates the pro-inflammatory cytokines interleukin-1 beta and -18, which are important mediators of inflammation. Abnormal activation of the inflammasome leads to many inflammatory diseases, including gout, silicosis, neurodegeneration, and genetically inherited periodic fever syndromes. Therefore, identification of small molecule inhibitors that target the inflammasome is an important step toward developing effective therapeutics for the treatment of inflammation. Here, we show that the herbal NF-kappaB inhibitory compound parthenolide inhibits the activity of multiple inflammasomes in macrophages by directly inhibiting the protease activity of caspase-1. Additional investigations of other NF-kappaB inhibitors revealed that the synthetic I kappaB kinase-beta inhibitor Bay 11-7082 and structurally related vinyl sulfone compounds selectively inhibit NLRP3 inflammasome activity in macrophages independent of their inhibitory effect on NF-kappaB activity. In vitro assays of the effect of parthenolide and Bay 11-7082 on the ATPase activity of NLRP3 demonstrated that both compounds inhibit the ATPase activity of NLRP3, suggesting that the inhibitory effect of these compounds on inflammasome activity could be mediated in part through their effect on the ATPase activity of NLRP3. Our results thus elucidate the molecular mechanism for the therapeutic anti-inflammatory activity of parthenolide and identify vinyl sulfones as a new class of potential therapeutics that target the NLRP3 inflammasome.


Asunto(s)
Antiinflamatorios/farmacología , Inflamación/tratamiento farmacológico , Nitrilos/farmacología , Sesquiterpenos/farmacología , Sulfonas/farmacología , Animales , Células de la Médula Ósea/metabolismo , Caspasa 1/metabolismo , Muerte Celular , Humanos , Immunoblotting , L-Lactato Deshidrogenasa/metabolismo , Macrófagos/metabolismo , Ratones , FN-kappa B/metabolismo , Sulfonas/química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...