Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 123
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Nat Immunol ; 15(8): 738-48, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24952504

RESUMEN

Assembly of the NLRP3 inflammasome activates caspase-1 and mediates the processing and release of the leaderless cytokine IL-1ß and thereby serves a central role in the inflammatory response and in diverse human diseases. Here we found that upon activation of caspase-1, oligomeric NLRP3 inflammasome particles were released from macrophages. Recombinant oligomeric protein particles composed of the adaptor ASC or the p.D303N mutant form of NLRP3 associated with cryopyrin-associated periodic syndromes (CAPS) stimulated further activation of caspase-1 extracellularly, as well as intracellularly after phagocytosis by surrounding macrophages. We found oligomeric ASC particles in the serum of patients with active CAPS but not in that of patients with other inherited autoinflammatory diseases. Our findings support a model whereby the NLRP3 inflammasome, acting as an extracellular oligomeric complex, amplifies the inflammatory response.


Asunto(s)
Proteínas Portadoras/inmunología , Caspasa 1/inmunología , Inflamasomas/inmunología , Inflamación/inmunología , Macrófagos/inmunología , Animales , Proteínas Reguladoras de la Apoptosis , Proteínas Adaptadoras de Señalización CARD , Proteínas Portadoras/sangre , Proteínas Portadoras/genética , Caspasa 1/genética , Caspasas/genética , Caspasas/inmunología , Caspasas Iniciadoras , Células Cultivadas , Síndromes Periódicos Asociados a Criopirina/sangre , Proteínas del Citoesqueleto/sangre , Proteínas del Citoesqueleto/genética , Proteínas del Citoesqueleto/inmunología , Células HEK293 , Humanos , Inflamasomas/sangre , Interleucina-1beta/sangre , Interleucina-1beta/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteína con Dominio Pirina 3 de la Familia NLR , Fagocitosis/inmunología , Transducción de Señal/inmunología
2.
Clin Immunol ; 249: 109287, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-36907540

RESUMEN

Sensorineural hearing loss is the most common type of hearing loss in adults and occurs due to damage of the inner ear caused by a range of factors including ageing, excessive noise, toxins, and cancer. Auto-inflammatory disease is also a cause of hearing loss and there is evidence that inflammation could contribute to hearing loss in other conditions. Within the inner ear there are resident macrophage cells that respond to insults and whose activation correlates with damage. The NLRP3 inflammasome is a multi-molecular pro-inflammatory protein complex that forms in activated macrophages and may contribute to hearing loss. The aim of this article is to discuss the evidence for the NLRP3 inflammasome and associated cytokines as potential therapeutic targets for sensorineural hearing loss in conditions ranging from auto-inflammatory disease to tumour-induced hearing loss in vestibular schwannoma.


Asunto(s)
Oído Interno , Pérdida Auditiva Sensorineural , Pérdida Auditiva , Adulto , Humanos , Inflamasomas/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Pérdida Auditiva Sensorineural/etiología , Pérdida Auditiva Sensorineural/metabolismo , Oído Interno/metabolismo , Pérdida Auditiva/complicaciones
3.
PLoS Genet ; 16(11): e1009084, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-33147210

RESUMEN

The liver possesses a remarkable regenerative capacity based partly on the ability of hepatocytes to re-enter the cell cycle and divide to replace damaged cells. This capability is substantially reduced upon chronic damage, but it is not clear if this is a cause or consequence of liver disease. Here, we investigate whether blocking hepatocyte division using two different mouse models affects physiology as well as clinical liver manifestations like fibrosis and inflammation. We find that in P14 Cdk1Liv-/- mice, where the division of hepatocytes is abolished, polyploidy, DNA damage, and increased p53 signaling are prevalent. Cdk1Liv-/- mice display classical markers of liver damage two weeks after birth, including elevated ALT, ALP, and bilirubin levels, despite the lack of exogenous liver injury. Inflammation was further studied using cytokine arrays, unveiling elevated levels of CCL2, TIMP1, CXCL10, and IL1-Rn in Cdk1Liv-/- liver, which resulted in increased numbers of monocytes. Ablation of CDK2-dependent DNA re-replication and polyploidy in Cdk1Liv-/- mice reversed most of these phenotypes. Overall, our data indicate that blocking hepatocyte division induces biological processes driving the onset of the disease phenotype. It suggests that the decrease in hepatocyte division observed in liver disease may not only be a consequence of fibrosis and inflammation, but also a pathological cue.


Asunto(s)
División Celular/fisiología , Hepatocitos/fisiología , Cirrosis Hepática/metabolismo , Animales , Apoptosis/fisiología , Proteína Quinasa CDC2/genética , Proteína Quinasa CDC2/metabolismo , Ciclo Celular , Quinasa 2 Dependiente de la Ciclina/genética , Quinasa 2 Dependiente de la Ciclina/metabolismo , Citocinas/inmunología , Citocinas/metabolismo , Modelos Animales de Enfermedad , Fibrosis/fisiopatología , Hepatitis/metabolismo , Hepatitis/fisiopatología , Hepatocitos/metabolismo , Inflamación/patología , Hígado/metabolismo , Hígado/patología , Cirrosis Hepática/genética , Cirrosis Hepática/patología , Masculino , Ratones , Ratones Noqueados , Transducción de Señal
4.
J Neurosci ; 41(13): 3025-3038, 2021 03 31.
Artículo en Inglés | MEDLINE | ID: mdl-33597269

RESUMEN

Alzheimer's disease is a devastating neurodegenerative disease with a dramatically increasing prevalence and no disease-modifying treatment. Inflammatory lifestyle factors increase the risk of developing Alzheimer's disease. Zinc deficiency is the most prevalent malnutrition in the world and may be a risk factor for Alzheimer's disease potentially through enhanced inflammation, although evidence for this is limited. Here we provide epidemiological evidence suggesting that zinc supplementation was associated with reduced risk and slower cognitive decline, in people with Alzheimer's disease and mild cognitive impairment. Using the APP/PS1 mouse model of Alzheimer's disease fed a control (35 mg/kg zinc) or diet deficient in zinc (3 mg/kg zinc), we determined that zinc deficiency accelerated Alzheimer's-like memory deficits without modifying amyloid ß plaque burden in the brains of male mice. The NLRP3-inflammasome complex is one of the most important regulators of inflammation, and we show here that zinc deficiency in immune cells, including microglia, potentiated NLRP3 responses to inflammatory stimuli in vitro, including amyloid oligomers, while zinc supplementation inhibited NLRP3 activation. APP/PS1 mice deficient in NLRP3 were protected against the accelerated cognitive decline with zinc deficiency. Collectively, this research suggests that zinc status is linked to inflammatory reactivity and may be modified in people to reduce the risk and slow the progression of Alzheimer's disease.SIGNIFICANCE STATEMENT Alzheimer's disease is a common condition mostly affecting the elderly. Zinc deficiency is also a global problem, especially in the elderly and also in people with Alzheimer's disease. Zinc deficiency contributes to many clinical disorders, including immune dysfunction. Inflammation is known to contribute to the risk and progression of Alzheimer's disease; thus, we hypothesized that zinc status would affect Alzheimer's disease progression. Here we show that zinc supplementation reduced the prevalence and symptomatic decline in people with Alzheimer's disease. In an animal model of Alzheimer's disease, zinc deficiency worsened cognitive decline because of an enhancement in NLRP3-driven inflammation. Overall, our data suggest that zinc status affects Alzheimer's disease progression, and that zinc supplementation could slow the rate of cognitive decline.


Asunto(s)
Enfermedad de Alzheimer/sangre , Disfunción Cognitiva/sangre , Progresión de la Enfermedad , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Zinc/sangre , Adulto , Anciano , Enfermedad de Alzheimer/diagnóstico por imagen , Enfermedad de Alzheimer/dietoterapia , Animales , Células Cultivadas , Disfunción Cognitiva/diagnóstico por imagen , Disfunción Cognitiva/dietoterapia , Suplementos Dietéticos , Femenino , Estudios de Seguimiento , Humanos , Mediadores de Inflamación/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Persona de Mediana Edad , Zinc/administración & dosificación , Zinc/deficiencia
5.
Glia ; 70(6): 1068-1083, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35150591

RESUMEN

Microglia, resident brain immune cells, are critical in orchestrating responses to central nervous system (CNS) injury. Many microglial functions, such as phagocytosis, motility and chemotaxis, are suggested to rely on chloride channels, including the volume-regulated anion channel (VRAC), but studies to date have relied on the use of pharmacological tools with limited specificity. VRAC has also been proposed as a drug target for acute CNS injury, and its role in microglial function is of considerable interest for developing CNS therapeutics. This study aimed to definitively confirm the contribution of VRAC in microglia function by using conditional LRRC8A-knockout mice, which lacked the essential VRAC subunit LRRC8A in microglia. We demonstrated that while VRAC contributed to cell volume regulation, it had no effect on phagocytic activity, cell migration or P2YR12-dependent chemotaxis. Moreover, loss of microglial VRAC did not affect microglial morphology or the extent of ischemic damage following stroke. We conclude that VRAC does not critically regulate microglial responses to brain injury and could be targetable in other CNS cell types (e.g., astrocytes) without impeding microglial function. Our results also demonstrate a role for VRAC in cell volume regulation but show that VRAC is not involved in several major cellular functions that it was previously thought to regulate, and point to other, alternative mechanisms of chloride transport in innate immunity.


Asunto(s)
Microglía , Accidente Cerebrovascular , Animales , Tamaño de la Célula , Transporte Iónico , Proteínas de la Membrana/metabolismo , Ratones , Microglía/metabolismo
6.
Glia ; 70(7): 1301-1316, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35353387

RESUMEN

The NLRP3 (NLR family, pyrin domain containing 3) inflammasome is a multi-protein complex responsible for the activation of caspase-1 and the subsequent cleavage and activation of the potent proinflammatory cytokines IL-1ß and IL-18, and pyroptotic cell death. NLRP3 is implicated as a driver of inflammation in a range of disorders including neurodegenerative diseases, type 2 diabetes, and atherosclerosis. A commonly reported mechanism contributing to NLRP3 inflammasome activation is potassium ion (K+ ) efflux across the plasma membrane. Identification of K+ channels involved in NLRP3 activation remains incomplete. Here, we investigated the role of the K+ channel THIK-1 in NLRP3 activation. Both pharmacological inhibitors and cells from THIK-1 knockout (KO) mice were used to assess THIK-1 contribution to macrophage NLRP3 activation in vitro. Pharmacological inhibition of THIK-1 inhibited caspase-1 activation and IL-1ß release from mouse bone-marrow-derived macrophages (BMDMs), mixed glia, and microglia in response to NLRP3 agonists. Similarly, BMDMs and microglia from THIK-1 KO mice had reduced NLRP3-dependent IL-1ß release in response to P2X7 receptor activation with ATP. Overall, these data suggest that THIK-1 is a regulator of NLRP3 inflammasome activation in response to ATP and identify THIK-1 as a potential therapeutic target for inflammatory disease.


Asunto(s)
Diabetes Mellitus Tipo 2 , Inflamasomas , Canales de Potasio de Dominio Poro en Tándem/metabolismo , Adenosina Trifosfato/metabolismo , Animales , Caspasa 1/metabolismo , Inflamasomas/metabolismo , Interleucina-1beta/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteína con Dominio Pirina 3 de la Familia NLR/genética , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Potasio/metabolismo , Canales de Potasio
7.
Immunology ; 165(4): 460-480, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35137954

RESUMEN

The NLRP3 inflammasome is a multiprotein complex that regulates caspase-1 activation and subsequent interleukin (IL)-1ß and IL-18 release from innate immune cells in response to infection or injury. Derivatives of the metabolites itaconate and fumarate, dimethyl itaconate (DMI), 4-octyl itaconate (4OI) and dimethyl fumarate (DMF) limit both expression and release of IL-1ß following NLRP3 inflammasome activation. However, the direct effects of these metabolite derivatives on NLRP3 inflammasome responses require further investigation. Using murine bone marrow-derived macrophages, mixed glia and organotypic hippocampal slice cultures (OHSCs), we demonstrate that DMI, 4OI and DMF pretreatments inhibit pro-inflammatory cytokine production in response to lipopolysaccharide (LPS), as well as inhibit subsequent NLRP3 inflammasome activation induced by nigericin. DMI, 4OI, DMF and monomethyl fumarate (MMF), another fumarate derivative, also directly inhibited biochemical markers of NLRP3 activation in LPS-primed macrophages, mixed glia, OHSCs and human macrophages in response to nigericin and imiquimod, including ASC speck formation, caspase-1 activation, gasdermin D cleavage and IL-1ß release. DMF, an approved treatment of multiple sclerosis, as well as DMI, 4OI and MMF, inhibited NLRP3 activation in macrophages in response to lysophosphatidylcholine, which is used to induce demyelination, suggesting a possible mechanism for DMF in multiple sclerosis through NLRP3 inhibition. The derivatives also reduced pro-IL-1α cleavage in response to the calcium ionophore ionomycin. Together, these findings reveal the immunometabolic regulation of both the priming and activation steps of NLRP3 activation in macrophages. Furthermore, we highlight itaconate and fumarate derivatives as potential therapeutic options in NLRP3- and IL-1α-driven diseases, including in the brain.


Asunto(s)
Inflamasomas , Esclerosis Múltiple , Animales , Caspasa 1/metabolismo , Caspasas/metabolismo , Fumaratos/metabolismo , Fumaratos/farmacología , Humanos , Inflamasomas/metabolismo , Interleucina-1beta/metabolismo , Lipopolisacáridos/farmacología , Macrófagos/metabolismo , Ratones , Esclerosis Múltiple/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Nigericina/farmacología , Succinatos
8.
Artif Organs ; 46(2): 259-272, 2022 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-34662442

RESUMEN

INTRODUCTION: Recent experimental evidence suggests normothermic machine perfusion of the vascularized composite allograft results in improved preservation compared to static cold storage, with less reperfusion injury in the immediate post-operative period. However, metabolic acidosis is a common feature of vascularized composite allograft perfusion, primarily due to the inability to process metabolic by-products. We evaluated the impact of combined limb-kidney perfusion on markers of metabolic acidosis and inflammation in a porcine model. METHODS: Ten paired pig forelimbs were used for this study, grouped as either limb-only (LO, n = 5) perfusion, or limb-kidney (LK, n = 5) perfusion. Infrared thermal imaging was used to determine homogeneity of perfusion. Lactate, bicarbonate, base, pH, and electrolytes, along with an inflammatory profile generated via the quantification of cytokines and cell-free DNA in the perfusate were recorded. RESULTS: The addition of a kidney to a limb perfusion circuit resulted in the rapid stabilization of lactate, bicarbonate, base, and pH. Conversely, the LO circuit became progressively acidotic, correlating in a significant increase in pro-inflammatory cytokines. Global perfusion across the limb was more homogenous with LK compared to LO. CONCLUSION: The addition of a kidney during limb perfusion results in significant improvements in perfusate biochemistry, with no evidence of metabolic acidosis.


Asunto(s)
Acidosis/prevención & control , Aloinjertos Compuestos , Riñón/fisiología , Perfusión/métodos , Animales , Miembro Anterior , Inflamación/prevención & control , Daño por Reperfusión , Sus scrofa
9.
Immunology ; 162(1): 84-91, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-32954500

RESUMEN

Excessive and dysregulated inflammation is known to contribute to disease progression. HSP90 is an intracellular chaperone known to regulate inflammatory processes including the NLRP3 inflammasome and secretion of the pro-inflammatory cytokine interleukin(IL)-1ß. Here, primarily using an in vitro inflammasome ASC speck assay, and an in vivo model of murine peritonitis, we tested the utility of HSP90 inhibitors as anti-inflammatory molecules. We report that the HSP90 inhibitor EC144 effectively inhibited inflammatory processes including priming and activation of NLRP3 in vitro and in vivo. A specific inhibitor of the ß HSP90 isoform was ineffective suggesting the importance of the α isoform in inflammatory signalling. EC144 inhibited IL-1ß and IL-6 in vivo when administered orally, and was brain-penetrant. These data suggest that HSP90 inhibitors may be useful for targeting inflammation in diverse diseases that are worsened by the presence of inflammation.


Asunto(s)
Proteínas HSP90 de Choque Térmico/antagonistas & inhibidores , Inflamasomas/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Animales , Citocinas/metabolismo , Inflamación/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Peritonitis/metabolismo , Isoformas de Proteínas/metabolismo , Transducción de Señal/fisiología
10.
PLoS Pathog ; 15(4): e1007694, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30978238

RESUMEN

Pulmonary infections are a major global cause of morbidity, exacerbated by an increasing threat from antibiotic-resistant pathogens. In this context, therapeutic interventions aimed at protectively modulating host responses, to enhance defence against infection, take on ever greater significance. Pseudomonas aeruginosa is an important multidrug-resistant, opportunistic respiratory pathogen, the clearance of which can be enhanced in vivo by the innate immune modulatory properties of antimicrobial host defence peptides from the cathelicidin family, including human LL-37. Initially described primarily as bactericidal agents, cathelicidins are now recognised as multifunctional antimicrobial immunomodulators, modifying host responses to pathogens, but the key mechanisms involved in these protective functions are not yet defined. We demonstrate that P. aeruginosa infection of airway epithelial cells promotes extensive infected cell internalisation of LL-37, in a manner that is dependent upon epithelial cell interaction with live bacteria, but does not require bacterial Type 3 Secretion System (T3SS). Internalised LL-37 acts as a second signal to induce inflammasome activation in airway epithelial cells, which, in contrast to myeloid cells, are relatively unresponsive to P. aeruginosa. We demonstrate that this is mechanistically dependent upon cathepsin B release, and NLRP3-dependent activation of caspase 1. These result in LL-37-mediated release of IL-1ß and IL-18 in a manner that is synergistic with P. aeruginosa infection, and can induce caspase 1-dependent death of infected epithelial cells, and promote neutrophil chemotaxis. We propose that cathelicidin can therefore act as a second signal, required by P. aeruginosa infected epithelial cells to promote an inflammasome-mediated altruistic cell death of infection-compromised epithelial cells and act as a "fire alarm" to enhance rapid escalation of protective inflammatory responses to an uncontrolled infection. Understanding this novel modulatory role for cathelicidins, has the potential to inform development of novel therapeutic strategies to antibiotic-resistant pathogens, harnessing innate immunity as a complementation or alternative to current interventions.


Asunto(s)
Péptidos Catiónicos Antimicrobianos/farmacología , Catelicidinas/farmacología , Células Epiteliales/inmunología , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Infecciones por Pseudomonas/inmunología , Pseudomonas aeruginosa/inmunología , Sistema Respiratorio/inmunología , Animales , Caspasa 1/metabolismo , Comunicación Celular , Células Cultivadas , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Humanos , Inmunidad Innata/efectos de los fármacos , Inmunidad Innata/inmunología , Inflamasomas/efectos de los fármacos , Inflamasomas/inmunología , Inflamasomas/metabolismo , Interleucina-18/genética , Interleucina-18/metabolismo , Interleucina-1beta/genética , Interleucina-1beta/metabolismo , Macrófagos Peritoneales/efectos de los fármacos , Macrófagos Peritoneales/inmunología , Macrófagos Peritoneales/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Proteína con Dominio Pirina 3 de la Familia NLR/genética , Neutrófilos/efectos de los fármacos , Neutrófilos/inmunología , Neutrófilos/metabolismo , Infecciones por Pseudomonas/tratamiento farmacológico , Infecciones por Pseudomonas/microbiología , Pseudomonas aeruginosa/efectos de los fármacos , Sistema Respiratorio/efectos de los fármacos , Sistema Respiratorio/metabolismo
11.
J Immunol ; 203(10): 2724-2734, 2019 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-31586037

RESUMEN

Alternatively activated macrophages are essential effector cells during type 2 immunity and tissue repair following helminth infections. We previously showed that Ym1, an alternative activation marker, can drive innate IL-1R-dependent neutrophil recruitment during infection with the lung-migrating nematode, Nippostrongylus brasiliensis, suggesting a potential role for the inflammasome in the IL-1-mediated innate response to infection. Although inflammasome proteins such as NLRP3 have important proinflammatory functions in macrophages, their role during type 2 responses and repair are less defined. We therefore infected Nlrp3 -/- mice with N. brasiliensis Unexpectedly, compared with wild-type (WT) mice, infected Nlrp3 -/- mice had increased neutrophilia and eosinophilia, correlating with enhanced worm killing but at the expense of increased tissue damage and delayed lung repair. Transcriptional profiling showed that infected Nlrp3 -/- mice exhibited elevated type 2 gene expression compared with WT mice. Notably, inflammasome activation was not evident early postinfection with N. brasiliensis, and in contrast to Nlrp3 -/- mice, antihelminth responses were unaffected in caspase-1/11-deficient or WT mice treated with the NLRP3-specific inhibitor MCC950. Together these data suggest that NLRP3 has a role in constraining lung neutrophilia, helminth killing, and type 2 immune responses in an inflammasome-independent manner.


Asunto(s)
Inflamasomas/fisiología , Enfermedades Pulmonares Parasitarias/inmunología , Proteína con Dominio Pirina 3 de la Familia NLR/fisiología , Nippostrongylus/inmunología , Infecciones por Strongylida/inmunología , Animales , Caspasa 1/fisiología , Quimiotaxis de Leucocito , Eosinofilia/etiología , Eosinofilia/inmunología , Furanos/farmacología , Compuestos Heterocíclicos de 4 o más Anillos , Inmunidad Innata , Indenos , Interleucina-4/farmacología , Lectinas/biosíntesis , Lectinas/genética , Pulmón/patología , Pulmón/fisiología , Enfermedades Pulmonares Parasitarias/complicaciones , Enfermedades Pulmonares Parasitarias/patología , Enfermedades Pulmonares Parasitarias/fisiopatología , Macrófagos Alveolares/enzimología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteína con Dominio Pirina 3 de la Familia NLR/antagonistas & inhibidores , Proteína con Dominio Pirina 3 de la Familia NLR/deficiencia , Proteína con Dominio Pirina 3 de la Familia NLR/genética , Neutrófilos/inmunología , Regeneración , Infecciones por Strongylida/complicaciones , Infecciones por Strongylida/patología , Infecciones por Strongylida/fisiopatología , Sulfonamidas/farmacología , Sulfonas , Transcripción Genética , beta-N-Acetilhexosaminidasas/biosíntesis , beta-N-Acetilhexosaminidasas/genética
12.
Proc Natl Acad Sci U S A ; 115(40): E9371-E9380, 2018 10 02.
Artículo en Inglés | MEDLINE | ID: mdl-30232264

RESUMEN

The NLRP3 inflammasome is an important regulator of inflammation and immunity. It is a multimolecular platform formed within cells that facilitates the activation of proinflammatory caspases to drive secretion of cytokines such as interleukin-1ß (IL-1ß). Knowledge of the mechanisms regulating formation of the NLRP3 inflammasome is incomplete. Here we report Cl- channel-dependent formation of dynamic ASC oligomers and inflammasome specks that remain inactive in the absence of K+ efflux. Formed after Cl- efflux exclusively, ASC specks are NLRP3 dependent, reversible, and inactive, although they further prime inflammatory responses, accelerating and enhancing release of IL-1ß in response to a K+ efflux-inducing stimulus. NEK7 is a specific K+ sensor and does not associate with NLRP3 under conditions stimulating exclusively Cl- efflux, but does after K+ efflux, activating the complex driving inflammation. Our investigation delivers mechanistic understanding into inflammasome activation and the regulation of inflammatory responses.


Asunto(s)
Proteínas Adaptadoras de Señalización CARD/metabolismo , Cloruros/metabolismo , Inflamasomas/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Multimerización de Proteína , Animales , Proteínas Adaptadoras de Señalización CARD/genética , Femenino , Inflamasomas/genética , Inflamación/genética , Inflamación/metabolismo , Inflamación/patología , Interleucina-1beta/genética , Interleucina-1beta/metabolismo , Transporte Iónico/genética , Masculino , Ratones , Ratones Noqueados , Quinasas Relacionadas con NIMA/genética , Quinasas Relacionadas con NIMA/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/genética , Potasio/metabolismo
13.
Proc Natl Acad Sci U S A ; 115(28): 7404-7409, 2018 07 10.
Artículo en Inglés | MEDLINE | ID: mdl-29954866

RESUMEN

Cerebral malaria (CM) is a serious neurological complication caused by Plasmodium falciparum infection. Currently, the only treatment for CM is the provision of antimalarial drugs; however, such treatment by itself often fails to prevent death or development of neurological sequelae. To identify potential improved treatments for CM, we performed a nonbiased whole-brain transcriptomic time-course analysis of antimalarial drug chemotherapy of murine experimental CM (ECM). Bioinformatics analyses revealed IL33 as a critical regulator of neuroinflammation and cerebral pathology that is down-regulated in the brain during fatal ECM and in the acute period following treatment of ECM. Consistent with this, administration of IL33 alongside antimalarial drugs significantly improved the treatment success of established ECM. Mechanistically, IL33 treatment reduced inflammasome activation and IL1ß production in microglia and intracerebral monocytes in the acute recovery period following treatment of ECM. Moreover, treatment with the NLRP3-inflammasome inhibitor MCC950 alongside antimalarial drugs phenocopied the protective effect of IL33 therapy in improving the recovery from established ECM. We further showed that IL1ß release from macrophages was stimulated by hemozoin and antimalarial drugs and that this was inhibited by MCC950. Our results therefore demonstrate that manipulation of the IL33-NLRP3 axis may be an effective therapy to suppress neuroinflammation and improve the efficacy of antimalarial drug treatment of CM.


Asunto(s)
Antimaláricos/farmacología , Encéfalo/parasitología , Sistemas de Liberación de Medicamentos/métodos , Interleucina-33/metabolismo , Malaria Cerebral/tratamiento farmacológico , Malaria Falciparum/tratamiento farmacológico , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Plasmodium falciparum/metabolismo , Animales , Encéfalo/metabolismo , Encéfalo/patología , Modelos Animales de Enfermedad , Femenino , Perfilación de la Expresión Génica , Hemoproteínas/metabolismo , Interleucina-1beta/biosíntesis , Interleucina-33/antagonistas & inhibidores , Macrófagos/metabolismo , Macrófagos/patología , Malaria Cerebral/metabolismo , Malaria Cerebral/patología , Malaria Falciparum/metabolismo , Malaria Falciparum/patología , Masculino , Ratones , Proteína con Dominio Pirina 3 de la Familia NLR/antagonistas & inhibidores , Transcriptoma/efectos de los fármacos
14.
J Biol Chem ; 294(21): 8325-8335, 2019 05 24.
Artículo en Inglés | MEDLINE | ID: mdl-30940725

RESUMEN

Interleukin (IL)-1 family cytokines potently regulate inflammation, with the majority of the IL-1 family proteins being secreted from immune cells via unconventional pathways. In many cases, secretion of IL-1 cytokines appears to be closely coupled to cell death, yet the secretory mechanisms involved remain poorly understood. Here, we studied the secretion of the three best-characterized members of the IL-1 superfamily, IL-1α, IL-1ß, and IL-18, in a range of conditions and cell types, including murine bone marrow-derived and peritoneal macrophages, human monocyte-derived macrophages, HeLa cells, and mouse embryonic fibroblasts. We discovered that IL-1ß and IL-18 share a common secretory pathway that depends upon membrane permeability and can operate in the absence of complete cell lysis and cell death. We also found that the pathway regulating the trafficking of IL-1α is distinct from the pathway regulating IL-1ß and IL-18. Although the release of IL-1α could also be dissociated from cell death, it was independent of the effects of the membrane-stabilizing agent punicalagin, which inhibited both IL-1ß and IL-18 release. These results reveal that in addition to their role as danger signals released from dead cells, IL-1 family cytokines can be secreted in the absence of cell death. We propose that models used in the study of IL-1 release should be considered context-dependently.


Asunto(s)
Células de la Médula Ósea/metabolismo , Interleucina-18/metabolismo , Interleucina-1alfa/metabolismo , Interleucina-1beta/metabolismo , Macrófagos Peritoneales/metabolismo , Animales , Células de la Médula Ósea/citología , Fibroblastos/citología , Fibroblastos/metabolismo , Células HeLa , Humanos , Taninos Hidrolizables/farmacología , Macrófagos Peritoneales/citología , Ratones , Transporte de Proteínas/efectos de los fármacos
15.
Immunology ; 161(1): 39-52, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32445196

RESUMEN

Microglial inflammation driven by the NACHT, LRR and PYD domain-containing protein 3 (NLRP3) inflammasome contributes to brain disease and is a therapeutic target. Most mechanistic studies on NLRP3 activation use two-dimensional pure microglial cell culture systems. Here we studied the activation of the NLRP3 inflammasome in organotypic hippocampal slices, which allowed us to investigate microglial NLRP3 activation in a three-dimensional, complex tissue architecture. Toll-like receptor 2 and 4 activation primed microglial inflammasome responses in hippocampal slices by increasing NLRP3 and interleukin-1ß expression. Nigericin-induced NLRP3 inflammasome activation was dynamically visualized in microglia through ASC speck formation. Downstream caspase-1 activation, gasdermin D cleavage, pyroptotic cell death and interleukin-1ß release were also detected, and these findings were consistent when using different NLRP3 stimuli such as ATP and imiquimod. NLRP3 inflammasome pathway inhibitors were effective in organotypic hippocampal slices. Hence, we have highlighted organotypic hippocampal slice culture as a valuable ex vivo tool to allow the future study of NLRP3 inflammasomes in a representative tissue section, aiding the discovery of further mechanistic insights and drug development.


Asunto(s)
Hipocampo/metabolismo , Interleucina-1beta/metabolismo , Microglía/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Animales , Antibacterianos/farmacología , Caspasa 1/metabolismo , Células Cultivadas , Activación Enzimática/fisiología , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Ratones , Ratones Endogámicos C57BL , Nigericina/farmacología , Técnicas de Cultivo de Órganos , Proteínas de Unión a Fosfato/metabolismo
16.
EMBO Rep ; 19(10)2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30206189

RESUMEN

The assembly and activation of the inflammasomes are tightly regulated by post-translational modifications, including ubiquitin. Deubiquitinases (DUBs) counteract the addition of ubiquitin and are essential regulators of immune signalling pathways, including those acting on the inflammasome. How DUBs control the assembly and activation of inflammasomes is unclear. Here, we show that the DUBs USP7 and USP47 regulate inflammasome activation in macrophages. Chemical inhibition of USP7 and USP47 blocks inflammasome formation, independently of transcription, by preventing ASC oligomerisation and speck formation. We also provide evidence that the ubiquitination status of NLRP3 itself is altered by inhibition of USP7 and USP47. Interestingly, we found that the activity of USP7 and USP47 increased in response to inflammasome activators. Using CRISPR/Cas9 in the macrophage cell line THP-1, we show that inflammasome activation is reduced when both USP7 and USP47 are knocked down. Altogether, these data reveal a new post-transcriptional role for USP47 and USP7 in inflammation by regulating inflammasome activation and the release of the pro-inflammatory cytokines IL-1ß and IL-18, and implicate dual USP7 and USP47 inhibitors as potential therapeutic agents for inflammatory disease.


Asunto(s)
Inflamación/genética , Proteína con Dominio Pirina 3 de la Familia NLR/genética , Ubiquitina Tiolesterasa/genética , Peptidasa Específica de Ubiquitina 7/genética , Sistemas CRISPR-Cas/genética , Enzimas Desubicuitinizantes/química , Enzimas Desubicuitinizantes/genética , Técnicas de Silenciamiento del Gen , Humanos , Inflamasomas/genética , Inflamasomas/metabolismo , Inflamación/patología , Interleucina-18/genética , Interleucina-1beta/genética , Macrófagos/metabolismo , Transducción de Señal/genética , Proteasas Ubiquitina-Específicas , Ubiquitinación/genética
17.
Molecules ; 25(23)2020 Nov 25.
Artículo en Inglés | MEDLINE | ID: mdl-33255820

RESUMEN

Inflammasomes are protein complexes which are important in several inflammatory diseases. Inflammasomes form part of the innate immune system that triggers the activation of inflammatory cytokines interleukin (IL)-1ß and IL-18. The inflammasome most studied in sterile inflammation and non-communicable disease is the NLRP3 inflammasome. Upon activation by diverse pathogen or disease associated signals, NLRP3 nucleates the oligomerization of an adaptor protein ASC forming a platform (the inflammasome) for the recruitment and activation of the protease caspase-1. Active caspase-1 catalyzes the processing and release of IL-1ß and IL-18, and via cleavage of the pore forming protein gasdermin D can drive pyroptotic cell death. This review focuses on the structural basis and mechanism for NLRP3 inflammasome signaling in the context of drug design, providing chemical structures, activities, and clinical potential of direct inflammasome inhibitors. A cryo-EM structure of NLRP3 bound to NEK7 protein provides structural insight and aids in the discovery of novel NLRP3 inhibitors utilizing ligand-based or structure-based approaches.


Asunto(s)
Descubrimiento de Drogas , Inflamasomas/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Animales , Ensayos Clínicos como Asunto , Diseño de Fármacos , Desarrollo de Medicamentos , Evaluación Preclínica de Medicamentos , Humanos , Inmunidad Innata , Inflamación/etiología , Inflamación/metabolismo , Modelos Moleculares , Quinasas Relacionadas con NIMA/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/química , Unión Proteica , Relación Estructura-Actividad
18.
Stroke ; 50(5): 1232-1239, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-31009361

RESUMEN

Background and Purpose- A major process contributing to cell death in the ischemic brain is inflammation. Inflammasomes are multimolecular protein complexes that drive inflammation through activation of proinflammatory cytokines, such as IL (interleukin)-1ß. Preclinical evidence suggests that IL-1ß contributes to a worsening of ischemic brain injury. Methods- Using a mouse middle cerebral artery thrombosis model, we examined the inflammatory response after stroke and the contribution of the NLRP3 (NACHT, LRR and PYD domains-containing protein 3) inflammasome to ischemic injury. Results- There was a marked inflammatory response after stroke characterized by increased expression of proinflammatory cytokines and NLRP3 and by recruitment of leukocytes to the injured tissue. Targeting NLRP3 with the inhibitor MCC950, or using mice in which NLRP3 was knocked out, had no effect on the extent of injury caused by stroke. Conclusions- These data suggest that the NLRP3 pathway does not contribute to the inflammation exacerbating ischemic brain damage, contradicting several recent reports to the contrary.


Asunto(s)
Lesiones Encefálicas/metabolismo , Isquemia Encefálica/metabolismo , Trombosis Intracraneal/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/deficiencia , Accidente Cerebrovascular/metabolismo , Animales , Lesiones Encefálicas/patología , Isquemia Encefálica/patología , Furanos/farmacología , Compuestos Heterocíclicos de 4 o más Anillos , Indenos , Inflamasomas/antagonistas & inhibidores , Inflamasomas/deficiencia , Trombosis Intracraneal/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteína con Dominio Pirina 3 de la Familia NLR/antagonistas & inhibidores , Accidente Cerebrovascular/patología , Sulfonamidas/farmacología , Sulfonas
19.
J Cell Sci ; 130(19): 3197-3202, 2017 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-28871048

RESUMEN

Extracellular proteins with important signalling roles in processes, such as inflammation and angiogenesis, are known to employ unconventional routes of protein secretion. Although mechanisms of unconventional protein secretion are beginning to emerge, the precise molecular details have remained elusive for the majority of cargo proteins secreted by unconventional means. Recent findings suggest that for two examples of unconventionally secreted proteins, interleukin 1ß (IL-1ß) and fibroblast growth factor 2 (FGF2), the common molecular principle of pore formation may be shared. Under specific experimental conditions, secretion of IL-1ß and FGF2 is triggered by phosphatidylinositol 4,5-bisphosphate [PI(4,5)P2]-dependent formation of pores across the plasma membrane. However, the underlying mechanisms are different, with FGF2 known to directly interact with PI(4,5)P2, whereas in the case of IL-1ß secretion, it is proposed that the N-terminal fragment of gasdermin D interacts with PI(4,5)P2 to form the pore. Thus, although implemented in different ways, these findings suggest that pore formation may be shared by the unconventional secretion mechanisms for FGF2 and IL-1ß in at least some cases. In this Opinion article, we discuss the unconventional mechanisms of FGF2 and IL-1ß release with a particular emphasis on recent discoveries suggesting the importance of pore formation on the plasma membrane.


Asunto(s)
Membrana Celular/metabolismo , Factor 2 de Crecimiento de Fibroblastos/metabolismo , Interleucina-1beta/metabolismo , Proteínas de Neoplasias/metabolismo , Vías Secretoras/fisiología , Animales , Humanos , Péptidos y Proteínas de Señalización Intracelular , Proteínas de Unión a Fosfato , Fosfatidilinositol 4,5-Difosfato/metabolismo
20.
Proc Natl Acad Sci U S A ; 112(13): 4050-5, 2015 Mar 31.
Artículo en Inglés | MEDLINE | ID: mdl-25775556

RESUMEN

Inflammation that contributes to acute cerebrovascular disease is driven by the proinflammatory cytokine interleukin-1 and is known to exacerbate resulting injury. The activity of interleukin-1 is regulated by multimolecular protein complexes called inflammasomes. There are multiple potential inflammasomes activated in diverse diseases, yet the nature of the inflammasomes involved in brain injury is currently unknown. Here, using a rodent model of stroke, we show that the NLRC4 (NLR family, CARD domain containing 4) and AIM2 (absent in melanoma 2) inflammasomes contribute to brain injury. We also show that acute ischemic brain injury is regulated by mechanisms that require ASC (apoptosis-associated speck-like protein containing a CARD), a common adaptor protein for several inflammasomes, and that the NLRP3 (NLR family, pyrin domain containing 3) inflammasome is not involved in this process. These discoveries identify the NLRC4 and AIM2 inflammasomes as potential therapeutic targets for stroke and provide new insights into how the inflammatory response is regulated after an acute injury to the brain.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/metabolismo , Lesiones Encefálicas/metabolismo , Proteínas de Unión al Calcio/metabolismo , Proteínas Portadoras/metabolismo , Proteínas de Unión al ADN/metabolismo , Regulación de la Expresión Génica , Animales , Encéfalo/metabolismo , Encéfalo/patología , Isquemia Encefálica/patología , Proteínas Adaptadoras de Señalización CARD , Muerte Celular , Citocinas/metabolismo , Hipoxia/patología , Infarto de la Arteria Cerebral Media/patología , Inflamación/patología , Isquemia/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Microscopía Fluorescente , Proteína con Dominio Pirina 3 de la Familia NLR , Estructura Terciaria de Proteína
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA