Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 98
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Immunity ; 44(4): 713-5, 2016 Apr 19.
Artículo en Inglés | MEDLINE | ID: mdl-27096310

RESUMEN

Generally, two signals are required to activate the NLRP3 inflammasome. In this issue of Immunity, Hornung and colleagues (2016) describe an alternative inflammasome activation pathway in human monocytes triggering activation of the NLRP3 inflammasome in response to a single stimulus.


Asunto(s)
Caspasa 1/metabolismo , Monocitos/metabolismo , Proteínas Portadoras/metabolismo , Humanos , Inflamasomas/metabolismo , Interleucina-1beta/metabolismo , Especies Reactivas de Oxígeno/metabolismo
2.
PLoS Pathog ; 18(8): e1010350, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-36044516

RESUMEN

Host-pathogen dynamics are constantly at play during enteroviral infection. Coxsackievirus B (CVB) is a common juvenile enterovirus that infects multiple organs and drives inflammatory diseases including acute pancreatitis and myocarditis. Much like other enteroviruses, CVB is capable of manipulating host machinery to hijack and subvert autophagy for its benefit. We have previously reported that CVB triggers the release of infectious extracellular vesicles (EVs) which originate from autophagosomes. These EVs facilitate efficient dissemination of infectious virus. Here, we report that TBK1 (Tank-binding kinase 1) suppresses release of CVB-induced EVs. TBK1 is a multimeric kinase that directly activates autophagy adaptors for efficient cargo recruitment and induces type-1 interferons during viral-mediated STING recruitment. Positioning itself at the nexus of pathogen elimination, we hypothesized that loss of TBK1 could exacerbate CVB infection due to its specific role in autophagosome trafficking. Here we report that infection with CVB during genetic TBK1 knockdown significantly increases viral load and potentiates the bulk release of viral EVs. Similarly, suppressing TBK1 with small interfering RNA (siRNA) caused a marked increase in intracellular virus and EV release, while treatment in vivo with the TBK1-inhibitor Amlexanox exacerbated viral pancreatitis and EV spread. We further demonstrated that viral EV release is mediated by the autophagy modifier proteins GABARAPL1 and GABARAPL2 which facilitate autophagic flux. We observe that CVB infection stimulates autophagy and increases the release of GABARAPL1/2-positive EVs. We conclude that TBK1 plays additional antiviral roles by inducing autophagic flux during CVB infection independent of interferon signaling, and the loss of TBK1 better allows CVB-laden autophagosomes to circumvent lysosomal degradation, increasing the release of virus-laden EVs. This discovery sheds new light on the mechanisms involved in viral spread and EV propagation during acute enteroviral infection and highlights novel intracellular trafficking protein targets for antiviral therapy.


Asunto(s)
Infecciones por Coxsackievirus , Enterovirus , Vesículas Extracelulares , Pancreatitis , Enfermedad Aguda , Proteínas Reguladoras de la Apoptosis/genética , Autofagia , Enterovirus/genética , Enterovirus Humano B/genética , Humanos , Proteínas Asociadas a Microtúbulos/genética , Proteínas Serina-Treonina Quinasas/genética , ARN Bicatenario , ARN Interferente Pequeño , Replicación Viral/genética
3.
Proc Natl Acad Sci U S A ; 118(1)2021 01 05.
Artículo en Inglés | MEDLINE | ID: mdl-33361152

RESUMEN

The balance between NLRP3 inflammasome activation and mitophagy is essential for homeostasis and cellular health, but this relationship remains poorly understood. Here we found that interleukin-1α (IL-1α)-deficient macrophages have reduced caspase-1 activity and diminished IL-1ß release, concurrent with reduced mitochondrial damage, suggesting a role for IL-1α in regulating this balance. LPS priming of macrophages induced pro-IL-1α translocation to mitochondria, where it directly interacted with mitochondrial cardiolipin (CL). Computational modeling revealed a likely CL binding motif in pro-IL-1α, similar to that found in LC3b. Thus, binding of pro-IL-1α to CL in activated macrophages may interrupt CL-LC3b-dependent mitophagy, leading to enhanced Nlrp3 inflammasome activation and more robust IL-1ß production. Mutation of pro-IL-1α residues predicted to be involved in CL binding resulted in reduced pro-IL-1α-CL interaction, a reduction in NLRP3 inflammasome activity, and increased mitophagy. These data identify a function for pro-IL-1α in regulating mitophagy and the potency of NLRP3 inflammasome activation.


Asunto(s)
Cardiolipinas/metabolismo , Interleucina-1alfa/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Animales , Autofagia , Cardiolipinas/fisiología , Caspasa 1/metabolismo , Femenino , Células HEK293 , Humanos , Inflamasomas/metabolismo , Interleucina-1alfa/fisiología , Macrófagos/metabolismo , Masculino , Ratones , Ratones Noqueados , Proteínas Asociadas a Microtúbulos/metabolismo , Mitocondrias/metabolismo , Mitofagia/fisiología , Proteína con Dominio Pirina 3 de la Familia NLR/fisiología , Unión Proteica/fisiología , Dominios Proteicos/fisiología , Especies Reactivas de Oxígeno/metabolismo
4.
Infect Immun ; 90(3): e0047021, 2022 03 17.
Artículo en Inglés | MEDLINE | ID: mdl-35130452

RESUMEN

Pseudomonas aeruginosa is a Gram-negative, opportunistic pathogen that causes nosocomial pneumonia, urinary tract infections, and bacteremia. A hallmark of P. aeruginosa pathogenesis is disruption of host cell function by the type III secretion system (T3SS) and its cognate exoenzyme effectors. The T3SS effector ExoU is phospholipase A2 (PLA2) that targets the host cell plasmalemmal membrane to induce cytolysis and is an important virulence factor that mediates immune avoidance. In addition, ExoU has been shown to subvert the host inflammatory response in a noncytolytic manner. In primary bone marrow-derived macrophages (BMDMs), P. aeruginosa infection is sensed by the nucleotide-binding domain containing leucine-rich repeats-like receptor 4 (NLRC4) inflammasome, which triggers caspase-1 activation and inflammation. ExoU transiently inhibits NLRC4 inflammasome-mediated activation of caspase-1 and its downstream target, interleukin 1ß (IL-1ß), to suppress activation of inflammation. In the present study, we sought to identify additional noncytolytic virulence functions for ExoU and discovered an unexpected association between ExoU, host mitochondria, and NLRC4. We show that infection of BMDMs with P. aeruginosa strains expressing ExoU elicited mitochondrial oxidative stress. In addition, mitochondria and mitochondrion-associated membrane fractions enriched from infected cells exhibited evidence of autophagy activation, indicative of damage. The observation that ExoU elicited mitochondrial stress and damage suggested that ExoU may also associate with mitochondria during infection. Indeed, ExoU phospholipase A2 enzymatic activity was present in enriched mitochondria and mitochondrion-associated membrane fractions isolated from P. aeruginosa-infected BMDMs. Intriguingly, enriched mitochondria and mitochondrion-associated membrane fractions isolated from infected Nlrc4 homozygous knockout BMDMs displayed significantly lower levels of ExoU enzyme activity, suggesting that NLRC4 plays a role in the ExoU-mitochondrion association. These observations prompted us to assay enriched mitochondria and mitochondrion-associated membrane fractions for NLRC4, caspase-1, and IL-1ß. NLRC4 and pro-caspase-1 were detected in enriched mitochondria and mitochondrion-associated membrane fractions isolated from noninfected BMDMs, and active caspase-1 and active IL-1ß were detected in response to P. aeruginosa infection. Interestingly, ExoU inhibited mitochondrion-associated caspase-1 and IL-1ß activation. The implications of ExoU-mediated effects on mitochondria and the NLRC4 inflammasome during P. aeruginosa infection are discussed.


Asunto(s)
Infecciones por Pseudomonas , Pseudomonas aeruginosa , Animales , Caspasa 1/metabolismo , Inflamasomas/metabolismo , Inflamación/metabolismo , Macrófagos/metabolismo , Ratones , Fosfolipasas/metabolismo , Pseudomonas aeruginosa/fisiología , Sistemas de Secreción Tipo III/metabolismo
5.
Immunity ; 39(2): 311-323, 2013 Aug 22.
Artículo en Inglés | MEDLINE | ID: mdl-23954133

RESUMEN

Nlrp3 inflammasome activation occurs in response to numerous agonists but the specific mechanism by which this takes place remains unclear. All previously evaluated activators of the Nlrp3 inflammasome induce the generation of mitochondrial reactive oxygen species (ROS), suggesting a model in which ROS is a required upstream mediator of Nlrp3 inflammasome activation. Here we have identified the oxazolidinone antibiotic linezolid as a Nlrp3 agonist that activates the Nlrp3 inflammasome independently of ROS. The pathways for ROS-dependent and ROS-independent Nlrp3 activation converged upon mitochondrial dysfunction and specifically the mitochondrial lipid cardiolipin. Cardiolipin bound to Nlrp3 directly and interference with cardiolipin synthesis specifically inhibited Nlrp3 inflammasome activation. Together these data suggest that mitochondria play a critical role in the activation of the Nlrp3 inflammasome through the direct binding of Nlrp3 to cardiolipin.


Asunto(s)
Cardiolipinas/metabolismo , Proteínas Portadoras/metabolismo , Inflamasomas/metabolismo , Mitocondrias/metabolismo , Acetamidas/metabolismo , Acetamidas/farmacología , Animales , Cardiolipinas/inmunología , Línea Celular , Ciclosporina/metabolismo , Activación Enzimática , Humanos , Inflamación/inducido químicamente , Interleucina-1beta/metabolismo , Interleucina-6/metabolismo , Linezolid , Macrófagos/inmunología , Macrófagos/metabolismo , Ratones , Mitocondrias/inmunología , Proteína con Dominio Pirina 3 de la Familia NLR , Oxazolidinonas/metabolismo , Oxazolidinonas/farmacología , Potasio/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal/inmunología , Factor de Necrosis Tumoral alfa/metabolismo
6.
Immunity ; 36(2): 155-7, 2012 Feb 24.
Artículo en Inglés | MEDLINE | ID: mdl-22365658

RESUMEN

Inhibitors of apoptosis proteins (IAPs) are important regulators of both cell death and inflammation. In this issue of Immunity, Vince et al. (2012) report that inhibition of IAPs results in the processing and secretion of IL-1ß through RIP3-mediated caspase-1- and caspase-8-dependent pathways.

7.
Hepatology ; 70(5): 1582-1599, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31044438

RESUMEN

Nonalcoholic fatty liver disease (NAFLD) enhances the growth and recurrence of colorectal cancer (CRC) liver metastasis. With the rising prevalence of NAFLD, a better understanding of the molecular mechanism underlying NAFLD-associated liver metastasis is crucial. Tumor-associated macrophages (TAMs) constitute a large portion of the tumor microenvironment that promotes tumor growth. NOD-like receptor C4 (NLRC4), a component of an inflammasome complex, plays a role in macrophage activation and interleukin (IL)-1ß processing. We aimed to investigate whether NLRC4-mediated TAM polarization contributes to metastatic liver tumor growth in NAFLD. Wild-type and NLRC4-/- mice were fed low-fat or high-fat diet for 6 weeks followed by splenic injection of mouse CRC MC38 cells. The tumors were analyzed 2 weeks after CRC cell injection. High-fat diet-induced NAFLD significantly increased the number and size of CRC liver metastasis. TAMs and CD206-expressing M2 macrophages accumulated markedly in tumors in the presence of NAFLD. NAFLD up-regulated the expression of IL-1ß, NLRC4, and M2 markers in tumors. In NAFLD, but not normal livers, deletion of NLRC4 decreased liver tumor growth accompanied by decreased M2 TAMs and IL-1ß expression in tumors. Wild-type mice showed increased vascularity and vascular endothelial growth factor (VEGF) expression in tumors with NAFLD, but these were reduced in NLRC4-/- mice. When IL-1 signaling was blocked by recombinant IL-1 receptor antagonist, liver tumor formation and M2-type macrophages were reduced, suggesting that IL-1 signaling contributes to M2 polarization and tumor growth in NAFLD. Finally, we found that TAMs, but not liver macrophages, produced more IL-1ß and VEGF following palmitate challenge. Conclusion: In NAFLD, NLRC4 contributes to M2 polarization, IL-1ß, and VEGF production in TAMs, which promote metastatic liver tumor growth.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/fisiología , Proteínas de Unión al Calcio/fisiología , Neoplasias del Colon/patología , Inflamasomas/fisiología , Neoplasias Hepáticas/complicaciones , Neoplasias Hepáticas/secundario , Enfermedad del Hígado Graso no Alcohólico/complicaciones , Animales , Femenino , Interleucina-1beta/fisiología , Macrófagos/patología , Ratones , Ratones Endogámicos C57BL
8.
J Immunol ; 200(9): 3047-3052, 2018 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-29602772

RESUMEN

The NLRP3 inflammasome is activated in response to microbial and danger signals, resulting in caspase-1-dependent secretion of the proinflammatory cytokines IL-1ß and IL-18. Canonical NLRP3 inflammasome activation is a two-step process requiring both priming and activation signals. During inflammasome activation, NLRP3 associates with mitochondria; however, the role for this interaction is unclear. In this article, we show that mouse NLRP3 and caspase-1 independently interact with the mitochondrial lipid cardiolipin, which is externalized to the outer mitochondrial membrane at priming in response to reactive oxygen species. An NLRP3 activation signal is then required for the calcium-dependent association of the adaptor molecule ASC with NLRP3 on the mitochondrial surface, resulting in inflammasome complex assembly and activation. These findings demonstrate a novel lipid interaction for caspase-1 and identify a role for mitochondria as supramolecular organizing centers in the assembly and activation of the NLRP3 inflammasome.


Asunto(s)
Cardiolipinas/metabolismo , Caspasa 1/metabolismo , Inflamasomas/metabolismo , Mitocondrias/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Animales , Cardiolipinas/inmunología , Caspasa 1/inmunología , Inflamasomas/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mitocondrias/inmunología , Proteína con Dominio Pirina 3 de la Familia NLR/inmunología
9.
J Immunol ; 200(3): 1188-1197, 2018 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-29282312

RESUMEN

Exaggerated inflammatory responses during influenza A virus (IAV) infection are typically associated with severe disease. Neutrophils are among the immune cells that can drive this excessive and detrimental inflammation. In moderation, however, neutrophils are necessary for optimal viral control. In this study, we explore the role of the nucleotide-binding domain leucine-rich repeat containing receptor family member Nlrp12 in modulating neutrophilic responses during lethal IAV infection. Nlrp12-/- mice are protected from lethality during IAV infection and show decreased vascular permeability, fewer pulmonary neutrophils, and a reduction in levels of neutrophil chemoattractant CXCL1 in their lungs compared with wild-type mice. Nlrp12-/- neutrophils and dendritic cells within the IAV-infected lungs produce less CXCL1 than their wild-type counterparts. Decreased CXCL1 production by Nlrp12-/- dendritic cells was not due to a difference in CXCL1 protein stability, but instead to a decrease in Cxcl1 mRNA stability. Together, these data demonstrate a previously unappreciated role for Nlrp12 in exacerbating the pathogenesis of IAV infection through the regulation of CXCL1-mediated neutrophilic responses.


Asunto(s)
Quimiocina CXCL1/metabolismo , Virus de la Influenza A/inmunología , Péptidos y Proteínas de Señalización Intracelular/genética , Infiltración Neutrófila/inmunología , Neutrófilos/inmunología , Infecciones por Orthomyxoviridae/inmunología , Animales , Permeabilidad Capilar/genética , Quimiocina CXCL1/genética , Células Dendríticas/inmunología , Femenino , Pulmón/inmunología , Pulmón/patología , Pulmón/virología , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Infecciones por Orthomyxoviridae/mortalidad , Infecciones por Orthomyxoviridae/virología , Estabilidad del ARN/genética , ARN Mensajero/genética
10.
Proc Natl Acad Sci U S A ; 114(27): E5444-E5453, 2017 07 03.
Artículo en Inglés | MEDLINE | ID: mdl-28630327

RESUMEN

Prostaglandin D2 (PGD2), an eicosanoid with both pro- and anti-inflammatory properties, is the most abundantly expressed prostaglandin in the brain. Here we show that PGD2 signaling through the D-prostanoid receptor 1 (DP1) receptor is necessary for optimal microglia/macrophage activation and IFN expression after infection with a neurotropic coronavirus. Genome-wide expression analyses indicated that PGD2/DP1 signaling is required for up-regulation of a putative inflammasome inhibitor, PYDC3, in CD11b+ cells in the CNS of infected mice. Our results also demonstrated that, in addition to PGD2/DP1 signaling, type 1 IFN (IFN-I) signaling is required for PYDC3 expression. In the absence of Pydc3 up-regulation, IL-1ß expression and, subsequently, mortality were increased in infected DP1-/- mice. Notably, survival was enhanced by IL1 receptor blockade, indicating that the effects of the absence of DP1 signaling on clinical outcomes were mediated, at least in part, by inflammasomes. Using bone marrow-derived macrophages in vitro, we confirmed that PYDC3 expression is dependent upon DP1 signaling and that IFN priming is critical for PYDC3 up-regulation. In addition, Pydc3 silencing or overexpression augmented or diminished IL-1ß secretion, respectively. Furthermore, DP1 signaling in human macrophages also resulted in the up-regulation of a putative functional analog, POP3, suggesting that PGD2 similarly modulates inflammasomes in human cells. These findings demonstrate a previously undescribed role for prostaglandin signaling in preventing excessive inflammasome activation and, together with previously published results, suggest that eicosanoids and inflammasomes are reciprocally regulated.


Asunto(s)
Coronavirus , Inflamasomas/metabolismo , Prostaglandina D2/metabolismo , Receptores de Prostaglandina/metabolismo , Transducción de Señal , Animales , AMP Cíclico/metabolismo , Citocinas/metabolismo , Humanos , Inflamación/metabolismo , Interferón Tipo I/metabolismo , Interleucina-1beta/metabolismo , Macrófagos/metabolismo , Ratones , Ratones Endogámicos C57BL , Análisis de Secuencia por Matrices de Oligonucleótidos , Dominios Proteicos , Receptores de Prostaglandina/antagonistas & inhibidores , Regulación hacia Arriba
11.
J Immunol ; 199(8): 2823-2833, 2017 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-28931602

RESUMEN

The role of the nucleotide-binding domain and leucine-rich repeat containing receptor NLRP10 in disease is incompletely understood. Using three mouse strains lacking the gene encoding NLRP10, only one of which had a coincidental mutation in DOCK8, we documented a role for NLRP10 as a suppressor of the cutaneous inflammatory response to Leishmania major infection. There was no evidence that the enhanced local inflammation was due to enhanced inflammasome activity. NLRP10/DOCK8-deficient mice harbored lower parasite burdens at the cutaneous site of inoculation compared with wild-type controls, whereas NLRP10-deficient mice and controls had similar parasite loads, suggesting that DOCK8 promotes local growth of parasites in the skin, whereas NLRP10 does not. NLRP10-deficient mice developed vigorous adaptive immune responses, indicating that there was not a global defect in the development of Ag-specific cytokine production. Bone marrow chimeras showed that the anti-inflammatory role of NLRP10 was mediated by NLRP10 expressed in resident cells in the skin rather than by bone marrow-derived cells. These data suggest a novel role for NLRP10 in the resolution of local inflammatory responses during L. major infection.


Asunto(s)
Antiinflamatorios/metabolismo , Proteínas Reguladoras de la Apoptosis/metabolismo , Leishmania major/inmunología , Leishmaniasis Cutánea/inmunología , Piel/inmunología , Proteínas Adaptadoras Transductoras de Señales , Animales , Proteínas Reguladoras de la Apoptosis/genética , Células Cultivadas , Citocinas/metabolismo , Femenino , Factores de Intercambio de Guanina Nucleótido/genética , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mutación/genética , Piel/parasitología
12.
Immunol Rev ; 265(1): 35-52, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25879282

RESUMEN

The NLRP3 (NOD-like receptor family, pyrin domain containing 3) inflammasome is a multiprotein complex that orchestrates innate immune responses to infection and cell stress through activation of caspase-1 and maturation of inflammatory cytokines pro-interleukin-1ß (pro-IL-1ß) and pro-IL-18. Activation of the inflammasome during infection can be protective, but unregulated NLRP3 inflammasome activation in response to non-pathogenic endogenous or exogenous stimuli can lead to unintended pathology. NLRP3 associates with mitochondria and mitochondrial molecules, and activation of the NLRP3 inflammasome in response to diverse stimuli requires cation flux, mitochondrial Ca(2+) uptake, and mitochondrial reactive oxygen species accumulation. It remains uncertain whether NLRP3 surveys mitochondrial integrity and senses mitochondrial damage, or whether mitochondria simply serve as a physical platform for inflammasome assembly. The structure of the active, caspase-1-processing NLRP3 inflammasome also requires further clarification, but recent studies describing the prion-like properties of ASC have advanced the understanding of how inflammasome assembly and caspase-1 activation occur while raising new questions regarding the propagation and resolution of NLRP3 inflammasome activation. Here, we review the mechanisms and pathways regulating NLRP3 inflammasome activation, discuss emerging concepts in NLRP3 complex organization, and expose the knowledge gaps hindering a comprehensive understanding of NLRP3 activation.


Asunto(s)
Proteínas Portadoras/metabolismo , Inflamasomas/metabolismo , Mitocondrias/metabolismo , Complejos Multiproteicos/metabolismo , Animales , Señalización del Calcio , Proteínas Portadoras/inmunología , Caspasa 1/metabolismo , Humanos , Inmunidad Innata , Inflamasomas/inmunología , Complejos Multiproteicos/inmunología , Proteína con Dominio Pirina 3 de la Familia NLR
13.
J Cell Sci ; 129(9): 1928-39, 2016 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-27006116

RESUMEN

Complement activation has long been associated with inflammation, primarily due to the elaboration of the complement anaphylotoxins C5a and C3a. In this work, we demonstrate that the phagocytosis of complement-opsonized particles promotes host inflammatory responses by a new mechanism that depends on the terminal complement components (C5b-C9). We demonstrate that during the phagocytosis of complement-opsonized particles, the membrane attack complex (MAC) of complement can be transferred from the activating particle to the macrophage plasma membrane by a 'bystander' mechanism. This MAC-mediated bystander damage initiates NLRP3 inflammasome activation, resulting in caspase-1 activation and IL-1ß and IL-18 secretion. Inflammasome activation is not induced when macrophages phagocytize unopsonized particles or particles opsonized with serum deficient in one of the terminal complement components. The secretion of IL-1ß and IL-18 by macrophages depends on NLRP3, ASC (also known as PYCARD) and caspase-1, as macrophages deficient in any one of these components fail to secrete these cytokines following phagocytosis. The phagocytosis of complement-opsonized particles increases leukocyte recruitment and promotes T helper 17 cell (TH17) biasing. These findings reveal a new mechanism by which complement promotes inflammation and regulates innate and adaptive immunity.


Asunto(s)
Efecto Espectador/inmunología , Complemento C3a/inmunología , Complemento C5a/inmunología , Complejo de Ataque a Membrana del Sistema Complemento/inmunología , Macrófagos/inmunología , Proteína con Dominio Pirina 3 de la Familia NLR/inmunología , Células Th17/inmunología , Animales , Proteínas Reguladoras de la Apoptosis/genética , Proteínas Reguladoras de la Apoptosis/inmunología , Efecto Espectador/genética , Proteínas Adaptadoras de Señalización CARD , Complemento C3a/genética , Complemento C5a/genética , Complejo de Ataque a Membrana del Sistema Complemento/genética , Células HEK293 , Humanos , Interleucina-18/genética , Interleucina-18/inmunología , Interleucina-1beta/genética , Interleucina-1beta/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Proteína con Dominio Pirina 3 de la Familia NLR/genética , Fagocitosis/genética
14.
PLoS Pathog ; 12(8): e1005803, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27482714

RESUMEN

Rickettsial agents are sensed by pattern recognition receptors but lack pathogen-associated molecular patterns commonly observed in facultative intracellular bacteria. Due to these molecular features, the order Rickettsiales can be used to uncover broader principles of bacterial immunity. Here, we used the bacterium Anaplasma phagocytophilum, the agent of human granulocytic anaplasmosis, to reveal a novel microbial surveillance system. Mechanistically, we discovered that upon A. phagocytophilum infection, cytosolic phospholipase A2 cleaves arachidonic acid from phospholipids, which is converted to the eicosanoid prostaglandin E2 (PGE2) via cyclooxygenase 2 (COX2) and the membrane associated prostaglandin E synthase-1 (mPGES-1). PGE2-EP3 receptor signaling leads to activation of the NLRC4 inflammasome and secretion of interleukin (IL)-1ß and IL-18. Importantly, the receptor-interacting serine/threonine-protein kinase 2 (RIPK2) was identified as a major regulator of the immune response against A. phagocytophilum. Accordingly, mice lacking COX2 were more susceptible to A. phagocytophilum, had a defect in IL-18 secretion and exhibited splenomegaly and damage to the splenic architecture. Remarkably, Salmonella-induced NLRC4 inflammasome activation was not affected by either chemical inhibition or genetic ablation of genes associated with PGE2 biosynthesis and signaling. This divergence in immune circuitry was due to reduced levels of the PGE2-EP3 receptor during Salmonella infection when compared to A. phagocytophilum. Collectively, we reveal the existence of a functionally distinct NLRC4 inflammasome illustrated by the rickettsial agent A. phagocytophilum.


Asunto(s)
Anaplasma phagocytophilum/inmunología , Proteínas Reguladoras de la Apoptosis/inmunología , Proteínas de Unión al Calcio/inmunología , Dinoprostona/inmunología , Ehrlichiosis/inmunología , Inflamasomas/inmunología , Subtipo EP3 de Receptores de Prostaglandina E/inmunología , Animales , Modelos Animales de Enfermedad , Ensayo de Inmunoadsorción Enzimática , Immunoblotting , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Reacción en Cadena en Tiempo Real de la Polimerasa
15.
Circ Res ; 119(10): 1101-1115, 2016 Oct 28.
Artículo en Inglés | MEDLINE | ID: mdl-27660287

RESUMEN

RATIONALE: Renal inflammation contributes to the pathophysiology of hypertension. CD161a+ immune cells are dominant in the (SHR) spontaneously hypertensive rat and expand in response to nicotinic cholinergic activation. OBJECTIVE: We aimed to phenotype CD161a+ immune cells in prehypertensive SHR after cholinergic activation with nicotine and determine if these cells are involved in renal inflammation and the development of hypertension. METHODS AND RESULTS: Studies used young SHR and WKY (Wistar-Kyoto) rats. Splenocytes and bone marrow cells were exposed to nicotine ex vivo, and nicotine was infused in vivo. Blood pressures, kidney, serum, and urine were obtained. Flow cytometry, Luminex/ELISA, immunohistochemistry, confocal microscopy, and Western blot were used. Nicotinic cholinergic activation induced proliferation of CD161a+/CD68+ macrophages in SHR-derived splenocytes, their renal infiltration, and premature hypertension in SHR. These changes were associated with increased renal expression of MCP-1 (monocyte chemoattractant protein-1) and VLA-4 (very-late antigen-4). LLT1 (lectin-like transcript 1), the ligand for CD161a, was overexpressed in SHR kidney, whereas vascular cellular and intracellular adhesion molecules were similar to those in WKY. Inflammatory cytokines were elevated in SHR kidney and urine after nicotine infusion. Nicotine-mediated renal macrophage infiltration/inflammation was enhanced in denervated kidneys, not explained by angiotensin II levels or expression of angiotensin type-1/2 receptors. Moreover, expression of the anti-inflammatory α7-nAChR (α7-nicotinic acetylcholine receptor) was similar in young SHR and WKY rats. CONCLUSIONS: A novel, inherited nicotinic cholinergic inflammatory effect exists in young SHR, measured by expansion of CD161a+/CD68+ macrophages. This leads to renal inflammation and premature hypertension, which may be partially explained by increased renal expression of LLT-1, MCP-1, and VLA-4.


Asunto(s)
Hipertensión/etiología , Riñón/patología , Macrófagos/efectos de los fármacos , Nicotina/farmacología , Edad de Inicio , Angiotensina II/metabolismo , Animales , Antígenos CD/análisis , Antígenos de Diferenciación Mielomonocítica/análisis , Movimiento Celular/efectos de los fármacos , Células Cultivadas , Quimiocina CCL2/biosíntesis , Quimiocina CCL2/genética , Citocinas/biosíntesis , Citocinas/genética , Desnervación , Regulación de la Expresión Génica/efectos de los fármacos , Hipertensión/genética , Hipertensión/metabolismo , Hipertensión/patología , Hipertensión Renal/etiología , Hipertensión Renal/genética , Hipertensión Renal/metabolismo , Hipertensión Renal/patología , Inmunofenotipificación , Integrina alfa4beta1/biosíntesis , Integrina alfa4beta1/genética , Riñón/inervación , Lectinas/biosíntesis , Lectinas/genética , Macrófagos/clasificación , Macrófagos/patología , Masculino , Subfamilia B de Receptores Similares a Lectina de Células NK/análisis , Nefritis/inducido químicamente , Nefritis/fisiopatología , Nicotina/toxicidad , Norepinefrina/metabolismo , Prehipertensión/etiología , Prehipertensión/genética , Prehipertensión/patología , Ratas , Ratas Endogámicas SHR , Ratas Endogámicas WKY , Receptor de Angiotensina Tipo 1/biosíntesis , Receptor de Angiotensina Tipo 1/genética , Receptor de Angiotensina Tipo 2/biosíntesis , Receptor de Angiotensina Tipo 2/genética , Receptor Nicotínico de Acetilcolina alfa 7/biosíntesis , Receptor Nicotínico de Acetilcolina alfa 7/genética
16.
Nature ; 484(7395): 510-3, 2012 Apr 25.
Artículo en Inglés | MEDLINE | ID: mdl-22538615

RESUMEN

NLRs (nucleotide-binding domain leucine-rich-repeat-containing receptors; NOD-like receptors) are a class of pattern recognition receptor (PRR) that respond to host perturbation from either infectious agents or cellular stress. The function of most NLR family members has not been characterized and their role in instructing adaptive immune responses remains unclear. NLRP10 (also known as PYNOD, NALP10, PAN5 and NOD8) is the only NLR lacking the putative ligand-binding leucine-rich-repeat domain, and has been postulated to be a negative regulator of other NLR members, including NLRP3 (refs 4-6). We did not find evidence that NLRP10 functions through an inflammasome to regulate caspase-1 activity nor that it regulates other inflammasomes. Instead, Nlrp10(-/-) mice had a profound defect in helper T-cell-driven immune responses to a diverse array of adjuvants, including lipopolysaccharide, aluminium hydroxide and complete Freund's adjuvant. Adaptive immunity was impaired in the absence of NLRP10 because of a dendritic cell (DC) intrinsic defect in emigration from inflamed tissues, whereas upregulation of DC costimulatory molecules and chemotaxis to CCR7-dependent and -independent ligands remained intact. The loss of antigen transport to the draining lymph nodes by a subset of migratory DCs resulted in an almost absolute loss in naive CD4(+) T-cell priming, highlighting the critical link between diverse innate immune stimulation, NLRP10 activity and the immune function of mature DCs.


Asunto(s)
Inmunidad Adaptativa/inmunología , Proteínas Reguladoras de la Apoptosis/metabolismo , Células Dendríticas/inmunología , Proteínas Adaptadoras Transductoras de Señales , Adyuvantes Inmunológicos , Animales , Antígenos/inmunología , Proteínas Reguladoras de la Apoptosis/deficiencia , Proteínas Reguladoras de la Apoptosis/genética , Proteínas Reguladoras de la Apoptosis/inmunología , Caspasa 1 , Movimiento Celular , Quimiocinas/inmunología , Células Dendríticas/citología , Células Dendríticas/metabolismo , Eliminación de Gen , Inflamasomas , Ligandos , Ganglios Linfáticos/inmunología , Ratones , Ratones Endogámicos BALB C , Linfocitos T/inmunología , Linfocitos T Colaboradores-Inductores/inmunología , Vacunas/inmunología
17.
Lung ; 196(6): 737-743, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30167842

RESUMEN

INTRODUCTION: Influenza infects millions of people each year causing respiratory distress and death in severe cases. On average, 200,000 people annually are hospitalized in the United States for influenza related complications. Tissue inhibitor of metalloproteinase-1 (TIMP-1), a secreted protein that inhibits MMPs, has been found to be involved in lung inflammation. Here, we evaluated the role of TIMP-1 in the host response to influenza-induced lung injury. METHODS: Wild-type (WT) and Timp1-deficient (Timp1-/-) mice that were 8-12 weeks old were administered A/PR/8/34 (PR8), a murine adapted H1N1 influenza virus, and euthanized 6 days after influenza installation. Bronchoalveolar lavage fluid and lungs were harvested from each mouse for ELISA, protein assay, PCR, and histological analysis. Cytospins were executed on bronchoalveolar lavage fluid to identify immune cells based on morphology and cell count. RESULTS: WT mice experienced significantly more weight loss compared to Timp1-/- mice after influenza infection. WT mice demonstrated more immune cell infiltrate and airway inflammation. Interestingly, PR8 levels were identical between the WT and Timp1-/- mice 6 days post-influenza infection. CONCLUSION: The data suggest that Timp1 promotes the immune response in the lungs after influenza infection facilitating an injurious phenotype as a result of influenza infection.


Asunto(s)
Lesión Pulmonar Aguda/inmunología , Lesión Pulmonar Aguda/metabolismo , Hemorragia/virología , Subtipo H1N1 del Virus de la Influenza A , Infecciones por Orthomyxoviridae/complicaciones , Inhibidor Tisular de Metaloproteinasa-1/genética , Inhibidor Tisular de Metaloproteinasa-1/metabolismo , Lesión Pulmonar Aguda/patología , Lesión Pulmonar Aguda/virología , Animales , Líquido del Lavado Bronquioalveolar/citología , Líquido del Lavado Bronquioalveolar/inmunología , Modelos Animales de Enfermedad , Recuento de Eritrocitos , Eritrocitos , Hemorragia/genética , Recuento de Leucocitos , Macrófagos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neutrófilos , Infecciones por Orthomyxoviridae/virología , Carga Viral/genética , Pérdida de Peso/genética
18.
Proc Natl Acad Sci U S A ; 112(10): 3056-61, 2015 Mar 10.
Artículo en Inglés | MEDLINE | ID: mdl-25713392

RESUMEN

Dendritic cells (DCs) are the primary leukocytes responsible for priming T cells. To find and activate naïve T cells, DCs must migrate to lymph nodes, yet the cellular programs responsible for this key step remain unclear. DC migration to lymph nodes and the subsequent T-cell response are disrupted in a mouse we recently described lacking the NOD-like receptor NLRP10 (NLR family, pyrin domain containing 10); however, the mechanism by which this pattern recognition receptor governs DC migration remained unknown. Using a proteomic approach, we discovered that DCs from Nlrp10 knockout mice lack the guanine nucleotide exchange factor DOCK8 (dedicator of cytokinesis 8), which regulates cytoskeleton dynamics in multiple leukocyte populations; in humans, loss-of-function mutations in Dock8 result in severe immunodeficiency. Surprisingly, Nlrp10 knockout mice crossed to other backgrounds had normal DOCK8 expression. This suggested that the original Nlrp10 knockout strain harbored an unexpected mutation in Dock8, which was confirmed using whole-exome sequencing. Consistent with our original report, NLRP3 inflammasome activation remained unaltered in NLRP10-deficient DCs even after restoring DOCK8 function; however, these DCs recovered the ability to migrate. Isolated loss of DOCK8 via targeted deletion confirmed its absolute requirement for DC migration. Because mutations in Dock genes have been discovered in other mouse lines, we analyzed the diversity of Dock8 across different murine strains and found that C3H/HeJ mice also harbor a Dock8 mutation that partially impairs DC migration. We conclude that DOCK8 is an important regulator of DC migration during an immune response and is prone to mutations that disrupt its crucial function.


Asunto(s)
Proteínas Portadoras/fisiología , Movimiento Celular/genética , Células Dendríticas/inmunología , Factores de Intercambio de Guanina Nucleótido/fisiología , Proteínas Adaptadoras Transductoras de Señales , Animales , Proteínas Reguladoras de la Apoptosis , Proteínas Portadoras/genética , Factores de Intercambio de Guanina Nucleótido/genética , Activación de Linfocitos , Ratones , Ratones Endogámicos C3H , Ratones Noqueados , Mutación Puntual
19.
J Infect Dis ; 216(9): 1164-1175, 2017 11 27.
Artículo en Inglés | MEDLINE | ID: mdl-28968905

RESUMEN

The Flavivirus genus within the Flaviviridae family is comprised of many important human pathogens including yellow fever virus (YFV), dengue virus (DENV), and Zika virus (ZKV), all of which are global public health concerns. Although the related flaviviruses hepatitis C virus and human pegivirus (formerly named GBV-C) interfere with T-cell receptor (TCR) signaling by novel RNA and protein-based mechanisms, the effect of other flaviviruses on TCR signaling is unknown. Here, we studied the effect of YFV, DENV, and ZKV on TCR signaling. Both YFV and ZKV replicated in human T cells in vitro; however, only YFV inhibited TCR signaling. This effect was mediated at least in part by the YFV envelope (env) protein coding RNA. Deletion mutagenesis studies demonstrated that expression of a short, YFV env RNA motif (vsRNA) was required and sufficient to inhibit TCR signaling. Expression of this vsRNA and YFV infection of T cells reduced the expression of a Src-kinase regulatory phosphatase (PTPRE), while ZKV infection did not. YFV infection in mice resulted in impaired TCR signaling and PTPRE expression, with associated reduction in murine response to experimental ovalbumin vaccination. Together, these data suggest that viruses within the flavivirus genus inhibit TCR signaling in a species-dependent manner.


Asunto(s)
Virus del Dengue/genética , ARN/genética , Receptores de Antígenos de Linfocitos T/genética , Transducción de Señal/genética , Replicación Viral/genética , Virus de la Fiebre Amarilla/genética , Virus Zika/genética , Virus del Dengue/patogenicidad , Humanos , Virus de la Fiebre Amarilla/patogenicidad , Virus Zika/patogenicidad
20.
Brain Behav Immun ; 62: 137-150, 2017 May.
Artículo en Inglés | MEDLINE | ID: mdl-28109896

RESUMEN

Both sleep loss and pathogens can enhance brain inflammation, sleep, and sleep intensity as indicated by electroencephalogram delta (δ) power. The pro-inflammatory cytokine interleukin-1 beta (IL-1ß) is increased in the cortex after sleep deprivation (SD) and in response to the Gram-negative bacterial cell-wall component lipopolysaccharide (LPS), although the exact mechanisms governing these effects are unknown. The nucleotide-binding domain and leucine-rich repeat protein-3 (NLRP3) inflammasome protein complex forms in response to changes in the local environment and, in turn, activates caspase-1 to convert IL-1ß into its active form. SD enhances the cortical expression of the somnogenic cytokine IL-1ß, although the underlying mechanism is, as yet, unidentified. Using NLRP3-gene knockout (KO) mice, we provide evidence that NLRP3 inflammasome activation is a crucial mechanism for the downstream pathway leading to increased IL-1ß-enhanced sleep. NLRP3 KO mice exhibited reduced non-rapid eye movement (NREM) sleep during the light period. We also found that sleep amount and intensity (δ activity) were drastically attenuated in NLRP3 KO mice following SD (homeostatic sleep response), as well as after LPS administration, although they were enhanced by central administration of IL-1ß. NLRP3, ASC, and IL1ß mRNA, IL-1ß protein, and caspase-1 activity were greater in the somatosensory cortex at the end of the wake-active period when sleep propensity was high and after SD in wild-type but not NLRP3 KO mice. Thus, our novel and converging findings suggest that the activation of the NLRP3 inflammasome can modulate sleep induced by both increased wakefulness and a bacterial component in the brain.


Asunto(s)
Inflamasomas/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Privación de Sueño/metabolismo , Sueño/fisiología , Animales , Inflamasomas/genética , Ratones , Ratones Noqueados , Proteína con Dominio Pirina 3 de la Familia NLR/genética , Polisomnografía , Transducción de Señal/fisiología , Privación de Sueño/genética , Vigilia/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA