Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
1.
Exp Neurol ; 346: 113856, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34474007

RESUMO

Vascular dementia (VaD) is the second most common form of dementia and is caused by vascular pathologies resulting in chronic cerebral hypoperfusion (CCH)- induced brain injury, and ultimately cognitive impairment and memory loss. Several lines of evidence have demonstrated chronic inflammation may be involved in VaD disease progression. It is now recognized that a major contributor to cerebral and systemic chronic inflammation involves the activation of innate immune molecular complexes termed inflammasomes. Whilst previous studies on animal models of VaD have focused on the cortex, hippocampus and striatum, few studies have investigated the effect of CCH on the cerebellum. Emerging studies have found new roles of the cerebellum in cognition, based on its structural interconnectivity with other brain regions and clinical relevance in neuropsychological deficits. In the present study, we conducted our investigation on the cerebellum using a CCH mouse model of VaD following bilateral common carotid artery stenosis (BCAS). This study is the first to characterize an increased expression of inflammasome receptors, adaptor and effector proteins, markers of inflammasome activation, proinflammatory cytokines, and apoptotic and pyroptotic cell death proteins in the cerebellum following CCH. Furthermore, in AIM2 knockout mice, we observed attenuated inflammasome-mediated production of proinflammatory cytokines, apoptosis, and pyroptosis in the cerebellum following CCH. Collectively, our findings provide novel evidence that AIM2 inflammasome activation promotes apoptosis and pyroptosis in the cerebellum following chronic hypoperfusion in a mouse model of VaD.


Assuntos
Apoptose/fisiologia , Lesões Encefálicas/metabolismo , Cerebelo/metabolismo , Proteínas de Ligação a DNA/metabolismo , Inflamassomos/metabolismo , Piroptose/fisiologia , Animais , Encéfalo/metabolismo , Encéfalo/patologia , Lesões Encefálicas/patologia , Morte Celular , Cerebelo/irrigação sanguínea , Cerebelo/patologia , Circulação Cerebrovascular/fisiologia , Proteínas de Ligação a DNA/antagonistas & inibidores , Proteínas de Ligação a DNA/deficiência , Inflamassomos/antagonistas & inibidores , Inflamassomos/deficiência , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout
2.
Front Immunol ; 12: 628168, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33717152

RESUMO

Obstructive sleep apnea (OSA) associated neurocognitive impairment is mainly caused by chronic intermittent hypoxia (CIH)-triggered neuroinflammation and oxidative stress. Previous study has demonstrated that mitochondrial reactive oxygen species (mtROS) was pivotal for hypoxia-related tissue injury. As a cytosolic multiprotein complex that participates in various inflammatory and neurodegenerative diseases, NLRP3 inflammasome could be activated by mtROS and thereby affected by the mitochondria-selective autophagy. However, the role of NLRP3 and possible mitophagy mechanism in CIH-elicited neuroinflammation remain to be elucidated. Compared with wild-type mice, NLRP3 deficiency protected them from CIH-induced neuronal damage, as indicated by the restoration of fear-conditioning test results and amelioration of neuron apoptosis. In addition, NLRP3 knockout mice displayed the mitigated microglia activation that elicited by CIH, concomitantly with elimination of damaged mitochondria and reduction of oxidative stress levels (malondialdehyde and superoxide dismutase). Elevated LC3 and beclin1 expressions were remarkably observed in CIH group. In vitro experiments, intermittent hypoxia (IH) significantly facilitated mitophagy induction and NLRP3 inflammasome activation in microglial (BV2) cells. Moreover, IH enhanced the accumulation of damaged mitochondria, increased mitochondrial depolarization and augmented mtROS release. Consistently, NLRP3 deletion elicited a protective phenotype against IH through enhancement of Parkin-mediated mitophagy. Furthermore, Parkin deletion or pretreated with 3MA (autophagy inhibitor) exacerbated these detrimental actions of IH, which was accompanied with NLRP3 inflammasome activation. These results revealed NLRP3 deficiency acted as a protective promotor through enhancing Parkin-depended mitophagy in CIH-induced neuroinflammation. Thus, NLRP3 gene knockout or pharmacological blockage could be as a potential therapeutic strategy for OSA-associated neurocognitive impairment.


Assuntos
Encéfalo/metabolismo , Inflamassomos/deficiência , Inflamação/prevenção & controle , Mitocôndrias/metabolismo , Mitofagia , Proteína 3 que Contém Domínio de Pirina da Família NLR/deficiência , Proteínas Quinases/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Síndromes da Apneia do Sono/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Animais , Encéfalo/imunologia , Encéfalo/patologia , Linhagem Celular , Modelos Animais de Doenças , Hipóxia/complicações , Inflamassomos/genética , Inflamação/imunologia , Inflamação/metabolismo , Inflamação/patologia , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microglia/imunologia , Microglia/metabolismo , Microglia/patologia , Mitocôndrias/imunologia , Mitocôndrias/patologia , Proteína 3 que Contém Domínio de Pirina da Família NLR/genética , Neuroimunomodulação , Estresse Oxidativo , Transdução de Sinais , Síndromes da Apneia do Sono/imunologia , Síndromes da Apneia do Sono/patologia
3.
Toxicol Appl Pharmacol ; 409: 115309, 2020 12 15.
Artigo em Inglês | MEDLINE | ID: mdl-33130049

RESUMO

The antimalarial drug artesunate (Art) has proven its beneficial effects against ischemia/reperfusion (I/R) injury in diverse organs, but its potential role against hepatic I/R is still obscure. This study, hence, examined whether treatment with Art alone or in combination with rapamycin (Rapa), an mTOR inhibitor, can ameliorate hepatic I/R injury via targeting the NLRP3 inflammasome signaling pathway. Rats were divided into hepatic sham- and I/R-operated rats. The latter were either left untreated (I/R group) or treated with Art, Rapa, or their combination. On the molecular level, all treatment regimens succeeded to hinder inflammasome assembly and activation, assessed as NLRP3, ASC, cleaved caspase-1, caspase-11, N-terminal cleaved gasdermin-D (GSDMD-N), IL-1ß, and IL-18. This effect was associated by the inhibition in the harmful signaling pathways HMGB1/RAGE and TLR4/MyD88/TRAF6 to inactivate the transcription factor NF-κB and the production of its pro-inflammatory cytokines IL-1ß, IL-18, IL-6, and TNF-α. Additionally, this effect entailed the inhibition of ICAM-1/MPO/ROS cascade, which in turn hampered cell demise induced by apoptosis, manifested as correction of the imbalanced Bcl2/Bax, as well as pyroptosis (LDH, cleaved caspase-1, caspase-11, GSDMD-N, IL-1ß, and IL-18), and necrosis. The corrected pathways were reflected on the improved liver function (serum ALT, AST, and LDH) and microscopical hepatic architecture. Noteworthy, the effect of Art on all parameters exceeded significantly that of Rapa and even improved the effect of the latter in the combination group. In conclusion, our results suggest novel roles for Art in abating functional and structural I/R-induced hepatic abnormalities via several traversing cross-talking pathways that succeeded to abate NLRP3 inflammasome and cell death.


Assuntos
Artesunato/farmacologia , Inflamassomos/deficiência , Hepatopatias/tratamento farmacológico , Fígado/efeitos dos fármacos , Estresse Oxidativo/efeitos dos fármacos , Traumatismo por Reperfusão/tratamento farmacológico , Animais , Apoptose/efeitos dos fármacos , Citocinas/metabolismo , Inflamassomos/metabolismo , Inflamação/metabolismo , Fígado/metabolismo , Hepatopatias/metabolismo , Masculino , Ratos , Ratos Wistar , Traumatismo por Reperfusão/metabolismo , Transdução de Sinais/efeitos dos fármacos
4.
Stroke ; 50(5): 1232-1239, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-31009361

RESUMO

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.


Assuntos
Lesões Encefálicas/metabolismo , Isquemia Encefálica/metabolismo , Trombose Intracraniana/metabolismo , Proteína 3 que Contém Domínio de Pirina da Família NLR/deficiência , Acidente Vascular Cerebral/metabolismo , Animais , Lesões Encefálicas/patologia , Isquemia Encefálica/patologia , Furanos/farmacologia , Compostos Heterocíclicos de 4 ou mais Anéis , Indenos , Inflamassomos/antagonistas & inibidores , Inflamassomos/deficiência , Trombose Intracraniana/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteína 3 que Contém Domínio de Pirina da Família NLR/antagonistas & inibidores , Acidente Vascular Cerebral/patologia , Sulfonamidas/farmacologia , Sulfonas
5.
Circulation ; 138(9): 898-912, 2018 08 28.
Artigo em Inglês | MEDLINE | ID: mdl-29588315

RESUMO

BACKGROUND: The CANTOS trial (Canakinumab Antiinflammatory Thrombosis Outcome Study) showed that antagonism of interleukin (IL)-1ß reduces coronary heart disease in patients with a previous myocardial infarction and evidence of systemic inflammation, indicating that pathways required for IL-1ß secretion increase cardiovascular risk. IL-1ß and IL-18 are produced via the NLRP3 inflammasome in myeloid cells in response to cholesterol accumulation, but mechanisms linking NLRP3 inflammasome activation to atherogenesis are unclear. The cholesterol transporters ATP binding cassette A1 and G1 (ABCA1/G1) mediate cholesterol efflux to high-density lipoprotein, and Abca1/g1 deficiency in myeloid cells leads to cholesterol accumulation. METHODS: To interrogate mechanisms connecting inflammasome activation with atherogenesis, we used mice with myeloid Abca1/g1 deficiency and concomitant deficiency of the inflammasome components Nlrp3 or Caspase-1/11. Bone marrow from these mice was transplanted into Ldlr-/- recipients, which were fed a Western-type diet. RESULTS: Myeloid Abca1/g1 deficiency increased plasma IL-18 levels in Ldlr-/- mice and induced IL-1ß and IL-18 secretion in splenocytes, which was reversed by Nlrp3 or Caspase-1/11 deficiency, indicating activation of the NLRP3 inflammasome. Nlrp3 or Caspase-1/11 deficiency decreased atherosclerotic lesion size in myeloid Abca1/g1-deficient Ldlr-/- mice. Myeloid Abca1/g1 deficiency enhanced caspase-1 cleavage not only in splenic monocytes and macrophages, but also in neutrophils, and dramatically enhanced neutrophil accumulation and neutrophil extracellular trap formation in atherosclerotic plaques, with reversal by Nlrp3 or Caspase-1/11 deficiency, suggesting that inflammasome activation promotes neutrophil recruitment and neutrophil extracellular trap formation in atherosclerotic plaques. These effects appeared to be indirectly mediated by systemic inflammation leading to activation and accumulation of neutrophils in plaques. Myeloid Abca1/g1 deficiency also activated the noncanonical inflammasome, causing increased susceptibility to lipopolysaccharide-induced mortality. Patients with Tangier disease, who carry loss-of-function mutations in ABCA1 and have increased myeloid cholesterol content, showed a marked increase in plasma IL-1ß and IL-18 levels. CONCLUSIONS: Cholesterol accumulation in myeloid cells activates the NLRP3 inflammasome, which enhances neutrophil accumulation and neutrophil extracellular trap formation in atherosclerotic plaques. Patients with Tangier disease, who have increased myeloid cholesterol content, showed markers of inflammasome activation, suggesting human relevance.


Assuntos
Transportador 1 de Cassete de Ligação de ATP/metabolismo , Membro 1 da Subfamília G de Transportadores de Cassetes de Ligação de ATP/metabolismo , Aterosclerose/prevenção & controle , Colesterol/metabolismo , Armadilhas Extracelulares/metabolismo , Inflamassomos/metabolismo , Inflamação/prevenção & controle , Células Mieloides/metabolismo , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Transportador 1 de Cassete de Ligação de ATP/deficiência , Transportador 1 de Cassete de Ligação de ATP/genética , Membro 1 da Subfamília G de Transportadores de Cassetes de Ligação de ATP/deficiência , Membro 1 da Subfamília G de Transportadores de Cassetes de Ligação de ATP/genética , Animais , Aterosclerose/genética , Aterosclerose/metabolismo , Aterosclerose/patologia , Estudos de Casos e Controles , Caspase 1/genética , Caspase 1/metabolismo , Caspases/genética , Caspases/metabolismo , Caspases Iniciadoras , Citocinas/sangue , Modelos Animais de Doenças , Humanos , Inflamassomos/deficiência , Inflamassomos/genética , Inflamação/genética , Inflamação/metabolismo , Inflamação/patologia , Camundongos Knockout , Células Mieloides/patologia , Proteína 3 que Contém Domínio de Pirina da Família NLR/deficiência , Proteína 3 que Contém Domínio de Pirina da Família NLR/genética , Placa Aterosclerótica , Receptores de LDL/genética , Receptores de LDL/metabolismo , Baço/metabolismo , Doença de Tangier/sangue , Doença de Tangier/genética
6.
Mol Neurobiol ; 53(7): 4874-82, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-26362308

RESUMO

Depression is a major public health concern in modern society, yet little is known about the molecular link between this condition and neuroinflammation. The inflammasome complex was recently shown to be implicated in depression. The present study shows the implication of NLRP3 inflammasome in animal model of stress-induced depression. Accordingly, we show here that in the absence of a NLRP3 inflammasome, prolonged stress does not provoke depressive behaviors or microglial activation in mice or dampen hippocampal neurogenesis. Indeed, NLRP3 deletion or inhibition of microglial activation impairs the stress-induced alterations associated with depression. According to these findings in animal model, the inflammasome could be a target for new therapeutic interventions to prevent depression in patients.


Assuntos
Depressão/metabolismo , Inflamassomos/deficiência , Proteína 3 que Contém Domínio de Pirina da Família NLR/deficiência , Estresse Psicológico/metabolismo , Animais , Depressão/patologia , Depressão/psicologia , Hipocampo/metabolismo , Hipocampo/patologia , Relações Interpessoais , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microglia/metabolismo , Microglia/patologia , Córtex Pré-Frontal/metabolismo , Córtex Pré-Frontal/patologia , Estresse Psicológico/patologia , Estresse Psicológico/psicologia
7.
Immunity ; 43(4): 751-63, 2015 Oct 20.
Artigo em Inglês | MEDLINE | ID: mdl-26384545

RESUMO

The crosstalk between inflammation and tumorigenesis is now clearly established. However, how inflammation is elicited in the metastatic environment and the corresponding contribution of innate immunity pathways in suppressing tumor growth at secondary sites are poorly understood. Here, we show that mice deficient in Nlrp3 inflammasome components had exacerbated liver colorectal cancer metastatic growth, which was mediated by impaired interleukin-18 (IL-18) signaling. Control of tumor growth was independent of differential cancer cell colonization or proliferation, intestinal microbiota effects, or tumoricidal activity by the adaptive immune system. Instead, the inflammasome-IL-18 pathway impacted maturation of hepatic NK cells, surface expression of the death ligand FasL, and capacity to kill FasL-sensitive tumors. Our results define a regulatory signaling circuit within the innate immune system linking inflammasome activation to effective NK-cell-mediated tumor attack required to suppress colorectal cancer growth in the liver.


Assuntos
Adenocarcinoma/secundário , Proteínas de Transporte/fisiologia , Neoplasias Colorretais/patologia , Inflamassomos/fisiologia , Células Matadoras Naturais/imunologia , Neoplasias Hepáticas/secundário , Adenocarcinoma/imunologia , Animais , Proteínas Reguladoras de Apoptose/deficiência , Proteínas de Ligação ao Cálcio/deficiência , Caspase 1/deficiência , Linhagem Celular Tumoral , Neoplasias Colorretais/imunologia , Citotoxicidade Imunológica , Proteínas de Ligação a DNA/deficiência , Proteína Ligante Fas/fisiologia , Microbioma Gastrointestinal , Imunidade Inata , Vigilância Imunológica , Inflamassomos/deficiência , Interleucina-18/fisiologia , Interleucina-1beta/fisiologia , Neoplasias Hepáticas/imunologia , Linfócitos do Interstício Tumoral/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteína 3 que Contém Domínio de Pirina da Família NLR , Proteínas de Neoplasias/deficiência , Proteínas de Neoplasias/fisiologia , Quimera por Radiação , Tolerância a Radiação , Microambiente Tumoral
8.
Methods Mol Biol ; 1040: 185-94, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23852605

RESUMO

Inflammasomes have emerged as central regulators of intestinal infection, immunity, and inflammation. Inflammasome activity mediates intestinal epithelial integrity, antimicrobial responses, and initiates inflammation through generation of the cytokines interleukin (IL-)1 and IL-18. Recent studies have identified an additional layer of inflammasome function in the intestine, namely, the control of intestinal microflora composition. Inflammasome-deficient mice show an aberrant microbial community which is dominantly transmissible to healthy mice. This dysbiosis in inflammasome-deficient mice has a profound impact on their physiology and pathophysiology, both locally in the intestine and systemically. Therefore, it is essential to consider the influence of the composition of microbial communities on experiments performed with inflammasome-deficient and other innate molecule-deficient mice, and to conduct experiments to control for potential dominant effects of the microflora on host responses. In this chapter, we provide experimental procedures to monitor inflammasome-mediated modifications of the intestinal microflora composition in mice and to test the resultant functional consequences of these changes in microbial communities and their transmission to cohoused mice.


Assuntos
Inflamassomos/deficiência , Camundongos Knockout/microbiologia , Microbiota , Animais , Intestinos/microbiologia , Metagenoma , Camundongos , Boca/microbiologia
9.
Exp Physiol ; 98(2): 462-72, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22848083

RESUMO

Heart failure is associated with a low-grade and chronic cardiac inflammation that impairs function; however, the mechanisms by which this sterile inflammation occurs in structural heart disease remain poorly defined. Cardiac-specific heterozygous overexpression of the calcineurin transgene (CNTg) in mice results in cardiac hypertrophy, inflammation, apoptosis and ventricular dilatation. We hypothesized that activation of the Nlrp3 inflammasome, an intracellular danger-sensing pathway required for processing the pro-inflammatory cytokine interleukin-1ß (IL-1ß), may contribute to myocardial dysfunction and disease progression. Here we report that Nlrp3 mRNA was increased in CNTg mice compared with wild-type. Consistent with inflammasome activation, CNTg animals had increased conversion of pro-caspase-1 to cleaved and activated forms, as well as markedly increased serum IL-1ß. Blockade of IL-1ß signalling via chronic IL-1 receptor antagonist therapy reduced cardiac inflammation and myocyte pathology in CNTg mice, resulting in improved systolic performance. Furthermore, genetic ablation of Nlrp3 in CNTg mice reduced pro-inflammatory cytokine maturation and cardiac inflammation, as well as improving systolic performance. These findings indicate that activation of the Nlrp3 inflammasome in CNTg mice promotes myocardial inflammation and systolic dysfunction through the production of pro-inflammatory IL-1ß. Blockade of IL-1ß signalling with the IL-1 receptor antagonist reverses these phenotypes and offers a possible therapeutic approach in the management of heart failure.


Assuntos
Cardiomiopatias/imunologia , Proteínas de Transporte/metabolismo , Insuficiência Cardíaca/imunologia , Inflamassomos/metabolismo , Mediadores da Inflamação/metabolismo , Interleucina-1beta/metabolismo , Miocardite/imunologia , Miocárdio/imunologia , Animais , Calcineurina/genética , Calcineurina/metabolismo , Cardiomiopatias/tratamento farmacológico , Cardiomiopatias/genética , Cardiomiopatias/patologia , Cardiomiopatias/fisiopatologia , Proteínas de Transporte/genética , Caspase 1/metabolismo , Modelos Animais de Doenças , Ativação Enzimática , Precursores Enzimáticos , Insuficiência Cardíaca/genética , Insuficiência Cardíaca/patologia , Insuficiência Cardíaca/fisiopatologia , Insuficiência Cardíaca/prevenção & controle , Inflamassomos/deficiência , Inflamassomos/genética , Mediadores da Inflamação/sangue , Proteína Antagonista do Receptor de Interleucina 1/farmacologia , Interleucina-1beta/sangue , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Miocardite/genética , Miocardite/patologia , Miocardite/fisiopatologia , Miocardite/prevenção & controle , Miocárdio/patologia , Células NIH 3T3 , Proteína 3 que Contém Domínio de Pirina da Família NLR , Receptores de Interleucina-1/antagonistas & inibidores , Receptores de Interleucina-1/metabolismo , Recuperação de Função Fisiológica , Transdução de Sinais , Sístole , Fatores de Tempo , Função Ventricular Esquerda
10.
Dig Dis ; 30 Suppl 1: 82-90, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23075874

RESUMO

BACKGROUND: Inflammatory bowel disease (IBD) is characterized by chronic intestinal inflammation due to dysregulation of the mucosal immune system. The cytokines IL-1ß and IL-18 appear early in intestinal inflammation and their pro-forms are processed via the caspase-1-activating multiprotein complex, the Nlrp3 inflammasome. Previously, we reported that the uptake of dextran sodium sulfate (DSS) by macrophages activates the Nlrp3 inflammasome and that Nlrp3(-/-) mice are protected in the acute DSS colitis model. Of note, other groups have reported opposing effects in regards to DSS susceptibility in Nlrp3(-/-) mice. Recently, mice lacking inflammasomes were found to develop a distinct intestinal microflora. METHODS: To reconcile the contradicting observations, we investigated the role of Nlrp3 deficiency in two different IBD models: acute DSS colitis and TNBS (2,4,6-trinitrobenzene sulfonic acid)-induced colitis. In addition, we investigated the impact of the intestinal flora on disease severity by performing cohousing experiments of wild-type and Nlrp3(-/-) mice, as well as by antibiotic treatment. RESULTS: Nlrp3(-/-) mice treated with either DSS or TNBS exhibited attenuated colitis and lower mortality. This protective effect correlated with an increased frequency of CD103+ lamina propria dendritic cells expressing a tolerogenic phenotype in Nlrp3(-/-) mice in steady state conditions. Interestingly, after cohousing, Nlrp3(-/-) mice were as susceptible as wild-type mice, indicating that transmission of endogenous bacterial flora between the two mouse strains might increase susceptibility of Nlrp3(-/-) mice towards DSS-induced colitis. Accordingly, treatment with antibiotics almost completely prevented colitis in the DSS model. CONCLUSIONS: The composition of the intestinal microflora significantly influences disease severity in IBD models comparing wild-type and Nlrp3(-/-) mice. This observation may - at least in part - explain contradictory results concerning the role of the inflammasome in different labs. Further studies are required to define the role of the Nlrp3 inflammasome in noninflamed mucosa under steady state conditions and in IBD.


Assuntos
Proteínas de Transporte/metabolismo , Meio Ambiente , Predisposição Genética para Doença , Inflamassomos/metabolismo , Doenças Inflamatórias Intestinais/genética , Doenças Inflamatórias Intestinais/imunologia , Substâncias Protetoras/metabolismo , Animais , Antibacterianos/farmacologia , Antibacterianos/uso terapêutico , Colite/imunologia , Colite/patologia , Colite/fisiopatologia , Células Dendríticas/imunologia , Modelos Animais de Doenças , Células Epiteliais/metabolismo , Células Epiteliais/patologia , Humanos , Tolerância Imunológica/imunologia , Inflamassomos/deficiência , Inflamassomos/genética , Doenças Inflamatórias Intestinais/tratamento farmacológico , Doenças Inflamatórias Intestinais/patologia , Intestinos/imunologia , Intestinos/microbiologia , Intestinos/patologia , Metagenoma , Camundongos , Mucosa/imunologia , Mucosa/patologia , Proteína 3 que Contém Domínio de Pirina da Família NLR , Fenótipo , Ácido Trinitrobenzenossulfônico
11.
Environ Health Perspect ; 120(12): 1692-8, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23010656

RESUMO

BACKGROUND: The role of the Nlrp3 inflammasome in nonallergic airway hyperresponsiveness (AHR) has not previously been reported. Recent evidence supports both interleukin (IL) 1ß and short fragments of hyaluronan (HA) as contributors to the biological response to inhaled ozone. OBJECTIVE: Because extracellular secretion of IL-1ß requires activation of the inflammasome, we investigated the role of the inflammasome proteins ASC, caspase1, and Nlrp3 in the biological response to ozone and HA. METHODS: C57BL/6J wild-type mice and mice deficient in ASC, caspase1, or Nlrp3 were exposed to ozone (1 ppm for 3 hr) or HA followed by analysis of airway resistance, cellular inflammation, and total protein and cytokines in bronchoalveolar lavage fluid (BALF). Transcription levels of IL-1ß and IL-18 were determined in two populations of lung macrophages. In addition, we examined levels of cleaved caspase1 and cleaved IL-1ß as markers of inflammasome activation in isolated alveolar macrophages harvested from BALF from HA-treated mice. RESULTS: We observed that genes of the Nlrp3 inflammasome were required for development of AHR following exposure to either ozone or HA fragments. These genes are partially required for the cellular inflammatory response to ozone. The expression of IL-1ß mRNA in alveolar macrophages was up-regulated after either ozone or HA challenge and was not dependent on the Nlrp3 inflammasome. However, soluble levels of IL-1ß protein were dependent on the inflammasome after challenge with either ozone or HA. HA challenge resulted in cleavage of macrophage-derived caspase1 and IL-1ß, suggesting a role for alveolar macrophages in Nlrp3-dependent AHR. CONCLUSIONS: The Nlrp3 inflammasome is required for the development of ozone-induced reactive airways disease.


Assuntos
Hiper-Reatividade Brônquica/induzido quimicamente , Proteínas de Transporte/metabolismo , Ácido Hialurônico/metabolismo , Inflamassomos/deficiência , Inflamassomos/metabolismo , Ozônio/toxicidade , Animais , Líquido da Lavagem Broncoalveolar/química , Caspase 1/metabolismo , Immunoblotting , Interleucina-1beta/metabolismo , Masculino , Cloreto de Metacolina/farmacologia , Camundongos , Camundongos Endogâmicos C57BL , Proteína 3 que Contém Domínio de Pirina da Família NLR , Reação em Cadeia da Polimerase em Tempo Real
12.
J Immunol ; 188(11): 5623-35, 2012 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-22547706

RESUMO

Bacterial flagellin is critical to mediate NLRC4 inflammasome-dependent caspase-1 activation. However, Shigella flexneri, a nonflagellated bacterium, and a flagellin (fliC) knockout strain of Pseudomonas aeruginosa are known to activate NLRC4 in bone marrow-derived macrophages. Furthermore, the flagellin-deficient fliC strain of P. aeruginosa was used in a mouse model of peritonitis to show the requirement of NLRC4. In a model of pulmonary P. aeruginosa infection, flagellin was shown to be essential for the induction of NLRC4-dependent caspase-1 activation. Moreover, in all P. aeruginosa studies, IL-1ß production was attenuated in NLRC4(-/-) mice; however, the role of IL-1ß in NLRC4-mediated innate immunity in the lungs against a nonflagellated bacterium was not explored. In this article, we report that NLRC4 is important for host survival and bacterial clearance, as well as neutrophil-mediated inflammation in the lungs following Klebsiella pneumoniae infection. NLRC4 is essential for K. pneumoniae-induced production of IL-1ß, IL-17A, and neutrophil chemoattractants (keratinocyte cell-derived chemokines, MIP-2, and LPS-induced CXC chemokines) in the lungs. NLRC4 signaling in hematopoietic cells contributes to K. pneumoniae-induced lung inflammation. Furthermore, exogenous IL-1ß, but not IL-18 or IL-17A, partially rescued survival, neutrophil accumulation, and cytokine/chemokine expression in the lungs of NLRC4(-/-) mice following infectious challenge. Furthermore, IL-1R1(-/-) mice displayed a decrease in neutrophilic inflammation in the lungs postinfection. Taken together, these findings provide novel insights into the role of NLRC4 in host defense against K. pneumoniae infection.


Assuntos
Proteínas Reguladoras de Apoptose/fisiologia , Proteínas de Ligação ao Cálcio/fisiologia , Inflamassomos/fisiologia , Interleucina-1beta/biossíntese , Infecções por Klebsiella/imunologia , Pneumonia Bacteriana/imunologia , Infecções Respiratórias/imunologia , Infecções Respiratórias/microbiologia , Animais , Proteínas Reguladoras de Apoptose/deficiência , Proteínas de Ligação ao Cálcio/deficiência , Caspase 1/metabolismo , Feminino , Humanos , Imunidade nas Mucosas/imunologia , Inflamassomos/deficiência , Mediadores da Inflamação/fisiologia , Infecções por Klebsiella/enzimologia , Infecções por Klebsiella/patologia , Macrófagos Alveolares/imunologia , Macrófagos Alveolares/microbiologia , Macrófagos Alveolares/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neutrófilos/imunologia , Neutrófilos/microbiologia , Neutrófilos/patologia , Pneumonia Bacteriana/microbiologia , Pneumonia Bacteriana/patologia , Infecções Respiratórias/patologia
13.
J Immunol ; 187(12): 6447-55, 2011 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-22079982

RESUMO

Although NLRC4/IPAF activation by flagellin has been extensively investigated, the downstream signaling pathways and the mechanisms responsible for infection clearance remain unclear. In this study, we used mice deficient for the inflammasome components in addition to wild-type (WT) Legionella pneumophila or bacteria deficient for flagellin (flaA) or motility (fliI) to assess the pathways responsible for NLRC4-dependent growth restriction in vivo and ex vivo. By comparing infections with WT L. pneumophila, fliI, and flaA, we found that flagellin and motility are important for the colonization of the protozoan host Acanthamoeba castellanii. However, in macrophages and mammalian lungs, flagellin expression abrogated bacterial replication. The flagellin-mediated growth restriction was dependent on NLRC4, and although it was recently demonstrated that NLRC4 is able to recognize bacteria independent of flagellin, we found that the NLRC4-dependent restriction of L. pneumophila multiplication was fully dependent on flagellin. By examining infected caspase-1(-/-) mice and macrophages with flaA, fliI, and WT L. pneumophila, we could detect greater replication of flaA, which suggests that caspase-1 only partially accounted for flagellin-dependent growth restriction. Conversely, WT L. pneumophila multiplied better in macrophages and mice deficient for NLRC4 compared with that in macrophages and mice deficient for caspase-1, supporting the existence of a novel caspase-1-independent response downstream of NLRC4. This response operated early after macrophage infection and accounted for the restriction of bacterial replication within bacteria-containing vacuoles. Collectively, our data indicate that flagellin is required for NLRC4-dependent responses to L. pneumophila and that NLRC4 triggers caspase-1-dependent and -independent responses for bacterial growth restriction in macrophages and in vivo.


Assuntos
Acanthamoeba castellanii/microbiologia , Proteínas Reguladoras de Apoptose/metabolismo , Proteínas de Ligação ao Cálcio/metabolismo , Proteínas de Transporte/fisiologia , Flagelos/imunologia , Legionella pneumophila/crescimento & desenvolvimento , Legionella pneumophila/imunologia , Macrófagos/imunologia , Macrófagos/microbiologia , Acanthamoeba castellanii/enzimologia , Acanthamoeba castellanii/imunologia , Animais , Proteínas Reguladoras de Apoptose/deficiência , Proteínas Reguladoras de Apoptose/genética , Carga Bacteriana/imunologia , Proteínas de Bactérias/genética , Células da Medula Óssea/enzimologia , Células da Medula Óssea/imunologia , Células da Medula Óssea/microbiologia , Proteínas de Ligação ao Cálcio/deficiência , Proteínas de Ligação ao Cálcio/genética , Proteínas de Transporte/genética , Linhagem Celular , Feminino , Flagelos/enzimologia , Flagelos/genética , Flagelina/biossíntese , Flagelina/genética , Inflamassomos/deficiência , Inflamassomos/genética , Legionella pneumophila/genética , Locomoção/imunologia , Macrófagos/enzimologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteína 3 que Contém Domínio de Pirina da Família NLR , ATPases Translocadoras de Prótons/genética , Transdução de Sinais/genética , Transdução de Sinais/imunologia
14.
J Immunol ; 187(2): 613-7, 2011 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-21677136

RESUMO

A common denominator among the multiple damage-inducing agents that ultimately lead to activation of NLRP3 has not yet been identified. Recently, production of reactive oxygen species (ROS) has been suggested to act as a common event upstream of the NLRP3 inflammasome machinery. Because de novo translation of NLRP3 is an essential step in the activation of NLRP3, we investigated the role of substances that inhibit either ROS production or its oxidative activity. Although we observe that NLRP3 inflammasome activation is unique among other known inflammasomes in its sensitivity to ROS inhibition, we have found that this phenomenon is attributable to the fact that NLRP3 strictly requires priming by a proinflammatory signal, a step that is blocked by ROS inhibitors. Although these data do not exclude a general role for ROS production in the process of NLRP3-triggered inflammation, they would put ROS upstream of NLRP3 induction, but not activation.


Assuntos
Proteínas de Transporte/antagonistas & inibidores , Regulação para Baixo/imunologia , Inflamassomos/antagonistas & inibidores , Inflamassomos/metabolismo , Ativação Linfocitária/imunologia , Espécies Reativas de Oxigênio/antagonistas & inibidores , Animais , Compostos de Bifenilo/farmacologia , Proteínas de Transporte/fisiologia , Células Cultivadas , Inflamassomos/deficiência , Macrófagos/imunologia , Macrófagos/metabolismo , Macrófagos/patologia , Glicoproteínas de Membrana , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Mutantes , NADPH Oxidase 2 , NADPH Oxidases , Proteína 3 que Contém Domínio de Pirina da Família NLR , Oniocompostos/farmacologia , Espécies Reativas de Oxigênio/metabolismo , Espécies Reativas de Oxigênio/farmacologia , Fator de Transcrição STAT1 , Transdução de Sinais/imunologia
15.
J Immunol ; 187(1): 64-73, 2011 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-21622869

RESUMO

IL-1ß is a cytokine critical to several inflammatory diseases in which pathogenic Th17 responses are implicated. Activation of the NLRP3 inflammasome by microbial and environmental stimuli can enable the caspase-1-dependent processing and secretion of IL-1ß. The acute-phase protein serum amyloid A (SAA) is highly induced during inflammatory responses, wherein it participates in systemic modulation of innate and adaptive immune responses. Elevated levels of IL-1ß, SAA, and IL-17 are present in subjects with severe allergic asthma, yet the mechanistic relationship among these mediators has yet to be identified. In this study, we demonstrate that Saa3 is expressed in the lungs of mice exposed to several mixed Th2/Th17-polarizing allergic sensitization regimens. SAA instillation into the lungs elicits robust TLR2-, MyD88-, and IL-1-dependent pulmonary neutrophilic inflammation. Furthermore, SAA drives production of IL-1α, IL-1ß, IL-6, IL-23, and PGE(2), causes dendritic cell (DC) maturation, and requires TLR2, MyD88, and the NLRP3 inflammasome for secretion of IL-1ß by DCs and macrophages. CD4(+) T cells polyclonally stimulated in the presence of conditioned media from SAA-exposed DCs produced IL-17, and the capacity of polyclonally stimulated splenocytes to secrete IL-17 is dependent upon IL-1, TLR2, and the NLRP3 inflammasome. Additionally, in a model of allergic airway inflammation, administration of SAA to the lungs functions as an adjuvant to sensitize mice to inhaled OVA, resulting in leukocyte influx after Ag challenge and a predominance of IL-17 production from restimulated splenocytes that is dependent upon IL-1R signaling.


Assuntos
Alérgenos/fisiologia , Proteínas de Transporte/metabolismo , Inflamassomos/metabolismo , Hipersensibilidade Respiratória/imunologia , Hipersensibilidade Respiratória/patologia , Proteína Amiloide A Sérica/fisiologia , Células Th17/imunologia , Células Th17/patologia , Alérgenos/genética , Animais , Proteínas de Transporte/genética , Polaridade Celular/genética , Polaridade Celular/imunologia , Células Cultivadas , Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Células Dendríticas/patologia , Modelos Animais de Doenças , Epitopos de Linfócito T/genética , Epitopos de Linfócito T/imunologia , Inflamassomos/deficiência , Inflamassomos/genética , Proteína Antagonista do Receptor de Interleucina 1/administração & dosagem , Interleucina-1alfa/antagonistas & inibidores , Interleucina-1alfa/fisiologia , Interleucina-1beta/metabolismo , Interleucina-1beta/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Proteína 3 que Contém Domínio de Pirina da Família NLR , Hipersensibilidade Respiratória/metabolismo , Mucosa Respiratória/imunologia , Mucosa Respiratória/metabolismo , Mucosa Respiratória/patologia , Transdução de Sinais/genética , Transdução de Sinais/imunologia , Baço/imunologia , Baço/metabolismo , Baço/patologia , Células Th17/metabolismo , Receptor 2 Toll-Like/deficiência , Receptor 2 Toll-Like/genética , Receptor 2 Toll-Like/fisiologia
16.
Gastroenterology ; 141(1): 358-69, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21439959

RESUMO

BACKGROUND & AIMS: Acute pancreatitis is characterized by early activation of intracellular proteases followed by acinar cell death and inflammation. Activation of damage-associated molecular pattern (DAMP) receptors and a cytosolic complex termed the inflammasome initiate forms of inflammation. In this study, we examined whether DAMP-receptors and the inflammasome provide the link between cell death and the initiation of inflammation in pancreatitis. METHODS: Acute pancreatitis was induced by caerulein stimulation in wild-type mice and mice deficient in components of the inflammasome (apoptosis-associated speck-like protein containing a caspase recruitment domain [ASC], NLRP3, caspase-1), Toll-like receptor 9 (TLR9), or the purinergic receptor P2X(7). Resident and infiltrating immune cell populations and pro-interleukin-1ß expression were characterized in control and caerulein-treated adult murine pancreas. TLR9 expression was quantified in pancreatic cell populations. Additionally, wild-type mice were pretreated with a TLR9 antagonist before induction of acute pancreatitis by caerulein or retrograde bile duct infusion of taurolithocholic acid 3-sulfate. RESULTS: Caspase-1, ASC, and NLRP3 were required for inflammation in acute pancreatitis. Genetic deletion of Tlr9 reduced pancreatic edema, inflammation, and pro-IL-1ß expression in pancreatitis. TLR9 was expressed in resident immune cells of the pancreas, which are predominantly macrophages. Pretreatment with the TLR9 antagonist IRS954 reduced pancreatic edema, inflammatory infiltrate, and apoptosis. Pretreatment with IRS954 reduced pancreatic necrosis and lung inflammation in taurolithocholic acid 3-sulfate-induced acute pancreatitis. CONCLUSIONS: Components of the inflammasome, ASC, caspase-1, and NLRP3, are required for the development of inflammation in acute pancreatitis. TLR9 and P2X(7) are important DAMP receptors upstream of inflammasome activation, and their antagonism could provide a new therapeutic strategy for treating acute pancreatitis.


Assuntos
Proteínas de Transporte/metabolismo , Inflamassomos/metabolismo , Pâncreas/imunologia , Pancreatite/imunologia , Transdução de Sinais , Receptor Toll-Like 9/metabolismo , Doença Aguda , Animais , Anti-Inflamatórios/farmacologia , Apoptose , Proteínas Reguladoras de Apoptose , Proteínas Adaptadoras de Sinalização CARD , Proteínas de Transporte/genética , Caspase 1/genética , Caspase 1/metabolismo , Ceruletídeo , Proteínas do Citoesqueleto/genética , Proteínas do Citoesqueleto/metabolismo , DNA/farmacologia , Modelos Animais de Doenças , Inflamassomos/deficiência , Inflamassomos/genética , Interleucina-1/metabolismo , Macrófagos/imunologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteína 3 que Contém Domínio de Pirina da Família NLR , Necrose , Infiltração de Neutrófilos , Pâncreas/efeitos dos fármacos , Pâncreas/patologia , Pancreatite/induzido quimicamente , Pancreatite/genética , Pancreatite/patologia , Pancreatite/prevenção & controle , Pneumonia/imunologia , Pneumonia/prevenção & controle , Precursores de Proteínas/metabolismo , Antagonistas do Receptor Purinérgico P2X/farmacologia , RNA Mensageiro/metabolismo , Receptores Purinérgicos P2X7/genética , Receptores Purinérgicos P2X7/metabolismo , Índice de Gravidade de Doença , Transdução de Sinais/efeitos dos fármacos , Ácido Taurolitocólico/análogos & derivados , Receptor Toll-Like 9/antagonistas & inibidores , Receptor Toll-Like 9/deficiência , Receptor Toll-Like 9/genética
17.
J Immunol ; 185(11): 7077-84, 2010 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-21037094

RESUMO

Aeromonas spp. are Gram-negative bacteria that cause serious infectious disease in humans. Such bacteria have been shown to induce apoptosis in infected macrophages, yet the host responses triggered by macrophage death are largely unknown. In this study, we demonstrate that the infection of mouse bone marrow-derived macrophages with Aeromonas veronii biotype sobria triggers activation of caspase-1 with the ensuing release of IL-1ß and pyroptosis. Caspase-1 activation in response to A. veronii infection requires the adaptor apoptosis-associated speck-like protein containing a caspase recruitment domain and both the NLRP3 and NLRC4 inflammasomes. Furthermore, caspase-1 activation requires aerolysin and a functional type III secretion system in A. veronii. Aerolysin-inducing caspase-1 activation is mediated through the NLRP3 inflammasome, with aerolysin-mediated cell death being largely dependent on the NLRP3 inflammasome. In contrast, the type III secretion system activates both the NLRP3 and NLRC4 inflammasomes. Inflammasome-mediated caspase-1 activation is also involved in host defenses against systemic A. veronii infection in mice. Our results indicated that multiple factors from both the bacteria and the host play a role in eliciting caspase-1 activation during A. veronii infection.


Assuntos
Aeromonas/imunologia , Proteínas Reguladoras de Apoptose/fisiologia , Sistemas de Secreção Bacterianos/imunologia , Proteínas de Ligação ao Cálcio/fisiologia , Proteínas de Transporte/fisiologia , Caspase 1/metabolismo , Inflamassomos/fisiologia , Proteínas Citotóxicas Formadoras de Poros/fisiologia , Aeromonas/patogenicidade , Animais , Proteínas Reguladoras de Apoptose/deficiência , Proteínas Reguladoras de Apoptose/genética , Toxinas Bacterianas/biossíntese , Células da Medula Óssea/imunologia , Células da Medula Óssea/microbiologia , Células da Medula Óssea/patologia , Proteínas de Ligação ao Cálcio/deficiência , Proteínas de Ligação ao Cálcio/genética , Proteínas de Transporte/genética , Caspase 1/deficiência , Caspase 1/genética , Morte Celular/genética , Morte Celular/imunologia , Células Cultivadas , Infecções por Bactérias Gram-Negativas/enzimologia , Infecções por Bactérias Gram-Negativas/genética , Infecções por Bactérias Gram-Negativas/imunologia , Inflamassomos/deficiência , Inflamassomos/genética , Macrófagos/imunologia , Macrófagos/microbiologia , Macrófagos/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteína 3 que Contém Domínio de Pirina da Família NLR , Proteínas Citotóxicas Formadoras de Poros/biossíntese
18.
J Neurosci ; 30(47): 15811-20, 2010 Nov 24.
Artigo em Inglês | MEDLINE | ID: mdl-21106820

RESUMO

Inflammation is increasingly recognized as an important contributor to a host of CNS disorders; however, its regulation in the brain is not well delineated. Nucleotide-binding domain, leucine-rich repeat, pyrin domain containing 3 (NLRP3) is a key component of the inflammasome complex, which also includes ASC (apoptotic speck-containing protein with a card) and procaspase-1. Inflammasome formation can be triggered by membrane P2X(7)R engagement leading to cleavage-induced maturation of caspase-1 and interleukin-1ß (IL-1ß)/IL-18. This work shows that expression of the Nlrp3 gene was increased >100-fold in a cuprizone-induced demyelination and neuroinflammation model. Mice lacking the Nlrp3 gene (Nlrp3(-/-)) exhibited delayed neuroinflammation, demyelination, and oligodendrocyte loss in this model. These mice also showed reduced demyelination in the experimental autoimmune encephalomyelitis model of neuroinflammation. This outcome is also observed for casp1(-/-) and IL-18(-/-) mice, whereas IL-1ß(-/-) mice were indistinguishable from wild-type controls, indicating that Nlrp3-mediated function is through caspase-1 and IL-18. Additional analyses revealed that, unlike the IL-1ß(-/-) mice, which have been previously shown to show delayed remyelination, Nlrp3(-/-) mice did not exhibit delayed remyelination. Interestingly, IL-18(-/-) mice showed enhanced remyelination, thus providing a possible compensatory mechanism for the lack of a remyelination defect in Nlrp3(-/-) mice. These results suggest that NLRP3 plays an important role in a model of multiple sclerosis by exacerbating CNS inflammation, and this is partly mediated by caspase-1 and IL-18. Additionally, the therapeutic inhibition of IL-18 might decrease demyelination but enhance remyelination, which has broad implications for demyelinating diseases.


Assuntos
Proteínas de Transporte/fisiologia , Caspase 1/fisiologia , Doenças Desmielinizantes/patologia , Encefalomielite Autoimune Experimental/patologia , Inflamassomos/fisiologia , Mediadores da Inflamação/fisiologia , Interleucina-18/fisiologia , Animais , Proteínas de Transporte/genética , Caspase 1/deficiência , Caspase 1/genética , Doenças Desmielinizantes/enzimologia , Doenças Desmielinizantes/metabolismo , Modelos Animais de Doenças , Encefalomielite Autoimune Experimental/enzimologia , Encefalomielite Autoimune Experimental/metabolismo , Inflamassomos/deficiência , Inflamassomos/genética , Mediadores da Inflamação/metabolismo , Interleucina-18/deficiência , Interleucina-18/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Esclerose Múltipla/enzimologia , Esclerose Múltipla/metabolismo , Esclerose Múltipla/patologia , Proteína 3 que Contém Domínio de Pirina da Família NLR , Transdução de Sinais/genética , Transdução de Sinais/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA