Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 87
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Nat Immunol ; 25(4): 693-702, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38486019

RESUMO

The inflammasome components NLRP3 and ASC are cytosolic proteins, which upon sensing endotoxins or danger cues, form multimeric complexes to process interleukin (IL)-1ß for secretion. Here we found that antigen (Ag)-triggered degranulation of IgE-sensitized mast cells (MCs) was mediated by NLRP3 and ASC. IgE-Ag stimulated NEK7 and Pyk2 kinases in MCs to induce the deposition of NLRP3 and ASC on granules and form a distinct protein complex (granulosome) that chaperoned the granules to the cell surface. MCs deficient in NLRP3 or ASC did not form granulosomes, degranulated poorly in vitro and did not evoke systemic anaphylaxis in mice. IgE-Ag-triggered anaphylaxis was prevented by an NLRP3 inhibitor. In endotoxin-primed MCs, pro-IL-1ß was rapidly packaged into granules after IgE-Ag stimulation and processed within granule remnants by proteases after degranulation, causing lethal anaphylaxis in mice. During IgE-Ag-mediated degranulation of endotoxin-primed MCs, granulosomes promoted degranulation, combined with exteriorization and processing of IL-1ß, resulting in severe inflammation.


Assuntos
Anafilaxia , Inflamassomos , Camundongos , Animais , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Mastócitos , Anafilaxia/metabolismo , Imunoglobulina E/metabolismo , Endotoxinas/metabolismo , Degranulação Celular
2.
Circulation ; 127(21): 2114-24, 2013 May 28.
Artigo em Inglês | MEDLINE | ID: mdl-23633271

RESUMO

BACKGROUND: CCR5 plays an important role in atherosclerosis and ischemic cardiovascular diseases, as well as in HIV replication and diffusion. HIV infection is characterized by a high burden of cardiovascular diseases, particularly in subjects exposed to ritonavir-boosted protease inhibitors. Maraviroc, a CCR5 antagonist antiretroviral drug, might provide benefit for patients with M-tropic HIV infections at high risk for cardiovascular diseases. METHODS AND RESULTS: Exposure to maraviroc limits the evolution and associated systemic inflammation of ritonavir-induced atherosclerotic in ApoE(-/-) mice and inhibits plaques development in a late model of atherosclerosis in which dyslipidemia plays the main pathogenic role. In ritonavir-treated mice, maraviroc reduced plaque areas and macrophage infiltration; downregulated the local expression of vascular cell adhesion molecule-1, intercellular adhesion molecule-1, monocyte chemoattractant protein-1, and interleukin-17A; and reduced tumor necrosis factor-α and RANTES (regulated on activation, normal T cell expressed, and secreted). Moreover, maraviroc counterregulated ritonavir-induced lipoatrophy and interlelukin-6 gene expression in epididymal fat, along with the splenic proinflammatory profile and expression of CD36 on blood monocytes. In the late model, maraviroc inhibited atherosclerotic progression by reducing macrophage infiltration and lowering the expression of adhesion molecules and RANTES inside the plaques. However, limited systemic inflammation was observed. CONCLUSIONS: In a mouse model of genetic dyslipidemia, maraviroc reduced the atherosclerotic progression by interfering with inflammatory cell recruitment into plaques. Moreover, in mice characterized by a general ritonavir-induced inflammation, maraviroc reversed the proinflammatory profile. Therefore, maraviroc could benefit HIV-positive patients with residual chronic inflammation who are at a high risk of acute coronary disease despite a suppressive antiretroviral therapy. To determine these benefits, large clinical studies are needed.


Assuntos
Antirretrovirais/efeitos adversos , Aterosclerose/induzido quimicamente , Aterosclerose/prevenção & controle , Antagonistas dos Receptores CCR5 , Cicloexanos/uso terapêutico , Placa Aterosclerótica/induzido quimicamente , Placa Aterosclerótica/prevenção & controle , Ritonavir/efeitos adversos , Triazóis/uso terapêutico , Animais , Apolipoproteínas E/deficiência , Apolipoproteínas E/genética , Aterosclerose/metabolismo , Movimento Celular , Quimiocina CCL2/metabolismo , Quimiocina CCL5/metabolismo , Modelos Animais de Doenças , Molécula 1 de Adesão Intercelular/metabolismo , Interleucina-17/metabolismo , Macrófagos/patologia , Masculino , Maraviroc , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Placa Aterosclerótica/metabolismo , Resultado do Tratamento , Fator de Necrose Tumoral alfa/metabolismo , Molécula 1 de Adesão de Célula Vascular/metabolismo
3.
Sci Immunol ; 9(93): eadi5578, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38427717

RESUMO

Urinary tract infections (UTIs) account for almost 25% of infections in women. Many are recurrent (rUTI), with patients frequently experiencing chronic pelvic pain and urinary frequency despite clearance of bacteriuria after antibiotics. To elucidate the basis for these bacteria-independent bladder symptoms, we examined the bladders of patients with rUTI. We noticed a notable increase in neuropeptide content in the lamina propria and indications of enhanced nociceptive activity. In mice subjected to rUTI, we observed sensory nerve sprouting that was associated with nerve growth factor (NGF) produced by recruited monocytes and tissue-resident mast cells. Treatment of rUTI mice with an NGF-neutralizing antibody prevented sprouting and alleviated pelvic sensitivity, whereas instillation of native NGF into naïve mice bladders mimicked nerve sprouting and pain behavior. Nerve activation, pain, and urinary frequency were each linked to the presence of proximal mast cells, because mast cell deficiency or treatment with antagonists against receptors of several direct or indirect mast cell products was each effective therapeutically. Thus, our findings suggest that NGF-driven sensory sprouting in the bladder coupled with chronic mast cell activation represents an underlying mechanism driving bacteria-independent pain and voiding defects experienced by patients with rUTI.


Assuntos
Mastócitos , Bexiga Urinária , Humanos , Camundongos , Feminino , Animais , Bexiga Urinária/inervação , Bexiga Urinária/metabolismo , Fator de Crescimento Neural/metabolismo , Reinfecção/complicações , Reinfecção/metabolismo , Dor/etiologia , Dor/metabolismo , Dor/prevenção & controle
4.
FASEB J ; 26(7): 3021-31, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22447981

RESUMO

The glucocorticoid receptor (GR) is a master gene orchestrating the activation of gluconeogenic genes in the liver in response to food withdrawal. Mechanisms of GR regulation by other nuclear receptors, however, are poorly defined. Here, we report that the farnesoid X receptor (FXR), a bile acid sensor, activates gluconeogenic pathways in the liver and regulates GR expression and activity. FXR-null mice are hypoglycemic in the unfed state and exhibit both a reduced hepatic production of glucose in response to the pyruvate challenge and a decreased expression of two rate-limiting enzymes involved in gluconeogenesis, phosphoenolpyruvate carboxykinase (PEPCK), and glucose-6-phosphatase (G6Pase), along with blunted liver expression of GR. Treating wild-type mice with a semisynthetic FXR ligand (6E-CDCA) increases the liver expression of GR, PEPCK, and G6Pase. This effect was lost in fed animals, as well as in FXR(-/-) mice. The human and mouse GR promoters contain a conserved FXR-responsive element (an ER-8 sequence) whose activation by FXR ligation leads to GR transcription. GR silencing by siRNA in vitro or its pharmacological antagonism in vivo with mifepristone reverses the effect of FXR activation on expression of gluconeogenic genes. These findings demonstrate that an FXR-GR pathway regulates the activation of hepatic gluconeogenesis in the transition from the unfed to the fed state.


Assuntos
Jejum/metabolismo , Gluconeogênese/fisiologia , Receptores Citoplasmáticos e Nucleares/metabolismo , Receptores de Glucocorticoides/metabolismo , Animais , Ácido Quenodesoxicólico/análogos & derivados , Ácido Quenodesoxicólico/metabolismo , Ácido Quenodesoxicólico/farmacologia , Gluconeogênese/genética , Glucose-6-Fosfatase/metabolismo , Células Hep G2 , Hepatócitos/metabolismo , Humanos , Ligantes , Fígado/efeitos dos fármacos , Fígado/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fosfoenolpiruvato Carboxiquinase (ATP)/metabolismo , Regiões Promotoras Genéticas , RNA Interferente Pequeno/genética , Receptores Citoplasmáticos e Nucleares/agonistas , Receptores Citoplasmáticos e Nucleares/deficiência , Receptores Citoplasmáticos e Nucleares/genética , Receptores de Glucocorticoides/antagonistas & inibidores , Receptores de Glucocorticoides/genética , Transdução de Sinais
5.
Pharmacol Res ; 77: 1-10, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-24004655

RESUMO

Glucocorticoids (GCs) are important endocrine regulators of a wide range of physiological processes ranging from immune function to glucose and lipid metabolism. For decades, synthetic glucocorticoids such as dexamethasone have been the cornerstone for the clinical treatment of inflammatory bowel diseases (IBD). A previous study has shown that farnesoid X receptor (FXR) enhances the transcription of NR3C1 gene, which encodes for human GR, by binding to a conserved FXR response element (FXRE) in the distal promoter of this gene. In the present study we demonstrate that FXR promotes the resolution of colitis in rodents by enhancing Gr gene transcription. We used the chromatin conformation capture (3C) assay to demonstrate that this FXRE is functional in mediating a head-to-tail chromatin looping, thus increasing Gr transcription efficiency. These findings underscore the importance of FXR/GR axis in the control of intestinal inflammation.


Assuntos
Cromatina/metabolismo , Colite/genética , Regiões Promotoras Genéticas/genética , Receptores Citoplasmáticos e Nucleares/metabolismo , Receptores de Glucocorticoides/genética , Animais , Anti-Inflamatórios/farmacologia , Anti-Inflamatórios/uso terapêutico , Células Cultivadas , Ácido Quenodesoxicólico/análogos & derivados , Ácido Quenodesoxicólico/antagonistas & inibidores , Ácido Quenodesoxicólico/farmacologia , Ácido Quenodesoxicólico/uso terapêutico , Cromatina/química , Cromatina/genética , Dexametasona/farmacologia , Dexametasona/uso terapêutico , Humanos , Camundongos , Camundongos Knockout , Mifepristona/farmacologia , Elementos de Resposta/genética , Ácido Trinitrobenzenossulfônico
6.
Mar Drugs ; 12(1): 36-53, 2013 Dec 24.
Artigo em Inglês | MEDLINE | ID: mdl-24368568

RESUMO

In the present study we provide evidence that solomonsterol A, a selective pregnane X receptor (PXR) agonist isolated from the marine sponge Theonella swinhoei, exerts anti-inflammatory activity and attenuates systemic inflammation and immune dysfunction in a mouse model of rheumatoid arthritis. Solomonsterol A was effective in protecting against the development of arthritis induced by injecting transgenic mice harboring a humanized PXR, with anti-collagen antibodies (CAIA) with beneficial effects on joint histopathology and local inflammatory response reducing the expression of inflammatory markers (TNFα, IFNγ and IL-17 and chemokines MIP1α and RANTES) in draining lymph nodes. Solomonsterol A rescued mice from systemic inflammation were assessed by measuring arthritis score, CRP and cytokines in the blood. In summary, the present study provides a molecular basis for the regulation of systemic local and systemic immunity by PXR agonists.


Assuntos
Anti-Inflamatórios , Artrite Reumatoide/tratamento farmacológico , Colanos/farmacologia , Síndromes de Imunodeficiência/tratamento farmacológico , Poríferos/química , Receptores de Esteroides/agonistas , Ésteres do Ácido Sulfúrico/farmacologia , Animais , Artrite Reumatoide/induzido quimicamente , Artrite Reumatoide/patologia , Proteína C-Reativa/metabolismo , Cartilagem/patologia , Quimiocina CCL3/metabolismo , Quimiocina CCL5/metabolismo , Colágeno Tipo II , Citocinas/sangue , Hepatócitos/efeitos dos fármacos , Humanos , Interferon gama/metabolismo , Interleucina-17/metabolismo , Fígado/efeitos dos fármacos , Fígado/metabolismo , Linfonodos/efeitos dos fármacos , Linfonodos/metabolismo , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Modelos Moleculares , Conformação Molecular , Receptor de Pregnano X , Receptores de Esteroides/biossíntese , Receptores de Esteroides/genética , Ativação Transcricional , Fator de Necrose Tumoral alfa/metabolismo
7.
Biochim Biophys Acta ; 1809(3): 157-65, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21296199

RESUMO

Multidrug resistance protein-4 (MRP4) is a member of the multidrug resistance associated gene family that is expressed on the basolateral membrane of hepatocytes and undergoes adaptive up-regulation in response to cholestatic injury or bile acid feeding. In this study we demonstrate that farnesoid X receptor (FXR) regulates MRP4 in vivo and in vitro. In vivo deletion of FXR induces MRP4 gene expression. In vitro treatment of HepG2 cells with FXR ligands, chenodeoxycholic acid (CDCA), cholic acid (CA) and the synthetic ligand GW-4064 suppresses basal mRNA level of the MRP4 gene as well as the co-treatment with CDCA and 6-(4-Chlorophenyl)imidazo[2,1-b][1,3]thiazole-5-carbaldehyde-O-(3,4-dichlorobenzyl)oxime (CITCO), an activator of constitutive androstane receptor (CAR). We found in the human MRP4 promoter a CAR responsive element (CARE) embedded within an FXR responsive element (FXRE). We cloned this region and found that FXR suppresses CAR activity in luciferase assay. Finally, we demonstrated that FXR competes with CAR for binding to this overlapping binding site. Our results support the view that FXR activation in obstructive cholestasis might worsen liver injury by hijacking a protective mechanism regulated by CAR and provides a new molecular explanation to the pathophysiology of cholestasis.


Assuntos
Proteínas Associadas à Resistência a Múltiplos Medicamentos/genética , Regiões Promotoras Genéticas , Receptores Citoplasmáticos e Nucleares/metabolismo , Animais , Sequência de Bases , Ácidos e Sais Biliares/farmacologia , Sítios de Ligação , Células Cultivadas , Receptor Constitutivo de Androstano , Regulação para Baixo/efeitos dos fármacos , Regulação para Baixo/genética , Deleção de Genes , Células Hep G2 , Hepatócitos/efeitos dos fármacos , Hepatócitos/metabolismo , Humanos , Cinética , Camundongos , Camundongos Endogâmicos C57BL , Modelos Biológicos , Dados de Sequência Molecular , Mutagênese/efeitos dos fármacos , Correpressor 1 de Receptor Nuclear/metabolismo , Ligação Proteica/efeitos dos fármacos , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Transcrição Gênica/efeitos dos fármacos
8.
Biochim Biophys Acta ; 1812(11): 1522-31, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21757002

RESUMO

Hepatic transport and metabolism of glutamate and glutamine are regulated by intervention of several proteins. Glutamine is taken up by periportal hepatocytes and is the major source of ammonia for urea synthesis and glutamate for N-acetylglutamate (NAG) synthesis, which is catalyzed by the N-acetylglutamate synthase (NAGS). Glutamate is taken up by perivenous hepatocytes and is the main source for the synthesis of glutamine, catalyzed by glutamine synthase (GS). Accumulation of glutamate and ammonia is a common feature of chronic liver failure, but mechanism that leads to failure of the urea cycle in this setting is unknown. The Farnesoid X Receptor (FXR) is a bile acid sensor in hepatocytes. Here, we have investigated its role in the regulation of the metabolism of both glutamine and glutamate. In vitro studies in primary cultures of hepatocytes from wild type and FXR(-/-) mice and HepG2 cells, and in vivo studies, in FXR(-/-) mice as well as in a rodent model of hepatic liver failure induced by carbon tetrachloride (CCl(4)), demonstrate a role for FXR in regulating this metabolism. Further on, promoter analysis studies demonstrate that both human and mouse NAGS promoters contain a putative FXRE, an ER8 sequence. EMSA, ChIP and luciferase experiments carried out to investigate the functionality of this sequence demonstrate that FXR is essential to induce the expression of NAGS. In conclusion, FXR activation regulates glutamine and glutamate metabolism and FXR ligands might have utility in the treatment of hyperammonemia states.


Assuntos
Glutamatos/metabolismo , Glutamina/metabolismo , Hiperamonemia/metabolismo , Cirrose Hepática Experimental/metabolismo , Receptores Citoplasmáticos e Nucleares/fisiologia , Aminoácido N-Acetiltransferase/genética , Aminoácido N-Acetiltransferase/metabolismo , Animais , Western Blotting , Tetracloreto de Carbono/toxicidade , Núcleo Celular , Imunoprecipitação da Cromatina , Ensaio de Desvio de Mobilidade Eletroforética , Células Hep G2 , Hepatócitos/citologia , Hepatócitos/metabolismo , Humanos , Hiperamonemia/induzido quimicamente , Cirrose Hepática Experimental/induzido quimicamente , Luciferases/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Regiões Promotoras Genéticas/genética , RNA Mensageiro/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa
9.
Biochim Biophys Acta ; 1802(3): 363-72, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-20060466

RESUMO

Farnesoid X Receptor plays an important role in maintaining bile acid, cholesterol homeostasis and glucose metabolism. Here we investigated whether FXR is expressed by pancreatic beta-cells and regulates insulin signaling in pancreatic beta-cell line and human islets. We found that FXR activation induces positive regulatory effects on glucose-induced insulin transcription and secretion by genomic and non-genomic activities. Genomic effects of FXR activation relay on the induction of the glucose regulated transcription factor KLF11. Indeed, results from silencing experiments of KLF11 demonstrate that this transcription factor is essential for FXR activity on glucose-induced insulin gene transcription. In addition FXR regulates insulin secretion by non-genomic effects. Thus, activation of FXR in betaTC6 cells increases Akt phosphorylation and translocation of the glucose transporter GLUT2 at plasma membrane, increasing the glucose uptake by these cells. In vivo experiments on Non Obese Diabetic (NOD) mice demonstrated that FXR activation delays development of signs of diabetes, hyperglycemia and glycosuria, by enhancing insulin secretion and by stimulating glucose uptake by the liver. These data established that an FXR-KLF11 regulated pathway has an essential role in the regulation of insulin transcription and secretion induced by glucose.


Assuntos
Insulina/genética , Insulina/metabolismo , Ilhotas Pancreáticas/metabolismo , Proteínas de Ligação a RNA/metabolismo , Transcrição Gênica , Animais , Proteínas Reguladoras de Apoptose , Western Blotting , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/patologia , Proteínas de Ciclo Celular/antagonistas & inibidores , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Células Cultivadas , Proteínas de Ligação a DNA/antagonistas & inibidores , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Imunofluorescência , Glucose/farmacologia , Transportador de Glucose Tipo 2/genética , Transportador de Glucose Tipo 2/metabolismo , Índice Glicêmico , Humanos , Técnicas Imunoenzimáticas , Secreção de Insulina , Insulinoma/genética , Insulinoma/metabolismo , Insulinoma/patologia , Ilhotas Pancreáticas/citologia , Neoplasias Hepáticas Experimentais/genética , Neoplasias Hepáticas Experimentais/metabolismo , Neoplasias Hepáticas Experimentais/patologia , Camundongos , Camundongos Endogâmicos NOD , Fosforilação , Proteínas Proto-Oncogênicas c-akt/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Proteínas de Ligação a RNA/antagonistas & inibidores , Proteínas de Ligação a RNA/genética , Proteínas Repressoras/antagonistas & inibidores , Proteínas Repressoras/genética , Proteínas Repressoras/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fatores de Transcrição/antagonistas & inibidores , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
10.
Inflamm Res ; 60(6): 577-87, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21279417

RESUMO

OBJECTIVE: The regulation of hepatic stellate cells (HSCs) by bacterial lipopolysaccharide (LPS) represents a recently-discovered and novel mechanism for hepatic injury and fibrosis. Stimulation of HSCs with LPS results in a rapid and marked induction of interleukin (IL)1ß, IL6 and tumor necrosis factor α. These events lead to the development of the activated phenotype in the HSCs associated with fibrosis and inflammation in the injured liver. We have previously demonstrated that farnesoid X receptor (FXR) activation increases transcription of rat peroxisome proliferator-activated receptor-γ (PPARγ) gene in HSCs. We aimed at evaluating the molecular mechanism of the transcriptional regulation of the PPARγ gene by FXR. METHODS: Real-time PCR, ELISA, transactivations, EMSA and ChIP experiments were performed in HSC-T6 cells, in primary HSCs, in HEK293T cells and in CCl(4)-treated rats. RESULTS: In vivo and in vitro activation of FXR downregulates cytokines and collagen(α)1 while inducing PPARγ and small heterodimer partner (SHP). NUBIScan analysis of rat PPARγ promoter revealed the presence of a putative FXR response element. Cotransfection with FXR/retinoic acid receptor significantly enhanced chenodeoxycholic acid-induced luciferase activity. EMSA experiments demonstrated that FXR was able to bind to an inverted repeat-1 sequence and ChIP experiments confirmed that FXR is recruited on the PPARγ promoter. CONCLUSION: The present study provides a molecular basis for the physiological cross-talk between FXR and PPARγ pathways in HSCs.


Assuntos
Doença Hepática Induzida por Substâncias e Drogas/metabolismo , Ácido Quenodesoxicólico/farmacologia , PPAR gama/genética , Receptores Citoplasmáticos e Nucleares/genética , Receptores Citoplasmáticos e Nucleares/metabolismo , Animais , Tetracloreto de Carbono , Linhagem Celular , Doença Hepática Induzida por Substâncias e Drogas/patologia , Regulação da Expressão Gênica/efeitos dos fármacos , Células HEK293 , Células Estreladas do Fígado/efeitos dos fármacos , Células Estreladas do Fígado/metabolismo , Humanos , Interleucina-1beta/metabolismo , Masculino , Miofibroblastos/efeitos dos fármacos , Miofibroblastos/metabolismo , Fenótipo , Ratos , Ratos Wistar , Fator de Necrose Tumoral alfa/metabolismo
11.
J Immunol ; 183(10): 6251-61, 2009 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-19864602

RESUMO

The farnesoid X receptor (FXR) is a bile acid-regulated nuclear receptor expressed in enterohepatic tissues. In this study we investigated whether FXR is expressed by cells of innate immunity and regulates inflammation in animal models of colitis. Acute (7 days) and chronic (8 wk) colitis were induced in wild-type and FXR(-/-) mice by intrarectal administration of trinitrobenzensulfonic acid or by 7-day administration of 5% dextran sulfate in drinking water. The results of this experiment demonstrate that FXR is expressed by and exerts counterregulatory effects on cells of innate immunity. Exposure of LPS-activated macrophages to 6-ethyl chenodeoxycholic acid (6E-CDCA; INT-747) a synthetic FXR ligand, results in a reciprocal regulation of NF-kappaB dependent-genes (TNF-alpha, IL-1beta, IL-6, COX-1, COX-2, and iNOS) and induction of SHP, a FXR-regulated gene. FXR activation stabilizes the nuclear corepressor NCoR on the NF-kappaB responsive element on the IL-1beta promoter. Colon inflammation in Crohn's disease patients and in rodent models of colitis is associated with a reduced expression of FXR mRNA. Using two rodent models of colon inflammation, we show that progression of these immune-mediated disorders is exacerbated in FXR(-/-) mice (p < 0.01). In vivo treatment with INT-747 attenuates organ injury and immune cell activation. FXR activation increased the colon expression of I-BABP, FXR, and SHP while reducing IL-1beta, IL-2, IL-6, TNF-alpha, and IFN-gamma mRNA expression and attenuating disease severity. In aggregate, these findings provide evidence that FXR is an essential component of a network of nuclear receptors that regulate intestinal innate immunity and homeostasis.


Assuntos
Colite/imunologia , Doença de Crohn/imunologia , Intestinos/imunologia , Macrófagos/imunologia , Receptores Citoplasmáticos e Nucleares/imunologia , Doença Aguda , Adulto , Animais , Doença Crônica , Colite/induzido quimicamente , Colite/metabolismo , Colite/patologia , Doença de Crohn/metabolismo , Feminino , Humanos , Imunidade Inata/efeitos dos fármacos , Imunidade Inata/imunologia , Interleucina-1beta/genética , Interleucina-1beta/imunologia , Interleucina-1beta/metabolismo , Mucosa Intestinal/metabolismo , Intestinos/efeitos dos fármacos , Intestinos/patologia , Lipopolissacarídeos/farmacologia , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos SCID , NF-kappa B/imunologia , NF-kappa B/metabolismo , Receptores Citoplasmáticos e Nucleares/agonistas , Receptores Citoplasmáticos e Nucleares/genética , Receptores Citoplasmáticos e Nucleares/metabolismo , Ácido Trinitrobenzenossulfônico/farmacologia
12.
J Immunol ; 183(10): 6657-66, 2009 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-19880446

RESUMO

Immune-mediated liver diseases including autoimmune and viral hepatitis are a major health problem worldwide. In this study, we report that activation of the farnesoid X receptor (FXR), a member of the ligand-activated nuclear receptor superfamily and bile sensor highly expressed in the liver, attenuates liver injury in a model of autoimmune hepatitis induced by Con A. We found that FXR gene ablation results in a time-dependent increase of liver expression (up to 20-fold in a 9-mo-old mouse) of osteopontin, a NKT cell-derived extracellular matrix protein and immunoregulatory cytokine. In comparison to wild-type, FXR(-/-) mice are more susceptible to Con A-induced hepatitis and react to Con A administration by an unregulated production of osteopontin. Administering wild-type mice with a synthetic FXR agonist attenuated Con A-induced liver damage and liver expression of the osteopontin gene. By in vitro studies, we found that FXR is expressed by primarily isolated NKT cells and its ablation favors ostepontin production in response to Con A. Chromatin immunoprecipitation assay and coimmunoprecipitation experiments demonstrate that the short heterodimer partner (SHP), a nuclear receptor and FXR target, was expressed by NKT cell hybridomas and increased in response to FXR activation. FXR activates SHP that interacts with and inhibits c-Jun binding to the osteopontin promoter. These data indicate that in NKT cells, FXR activation causes a SHP-mediated inhibition of osteopontin production. These data support the notion that the bile acid sensor FXR regulates the activation of liver NKT cells.


Assuntos
Hepatite/imunologia , Células T Matadoras Naturais/imunologia , Osteopontina/imunologia , Receptores Citoplasmáticos e Nucleares/imunologia , Doença Aguda , Animais , Ácidos e Sais Biliares/imunologia , Ácidos e Sais Biliares/metabolismo , Linhagem Celular , Concanavalina A/farmacologia , Hepatite/genética , Hepatite/metabolismo , Interferon gama/agonistas , Interferon gama/imunologia , Interferon gama/metabolismo , Interleucina-4/agonistas , Interleucina-4/imunologia , Interleucina-4/metabolismo , Fígado/imunologia , Fígado/patologia , Ativação Linfocitária/efeitos dos fármacos , Ativação Linfocitária/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mitógenos/farmacologia , Células T Matadoras Naturais/metabolismo , Osteopontina/agonistas , Osteopontina/metabolismo , Regiões Promotoras Genéticas/efeitos dos fármacos , Regiões Promotoras Genéticas/imunologia , RNA Mensageiro/efeitos dos fármacos , RNA Mensageiro/imunologia , RNA Mensageiro/metabolismo , Receptores Citoplasmáticos e Nucleares/genética , Receptores Citoplasmáticos e Nucleares/metabolismo , Fator de Necrose Tumoral alfa/agonistas , Fator de Necrose Tumoral alfa/imunologia , Fator de Necrose Tumoral alfa/metabolismo
13.
J Lipid Res ; 51(4): 771-84, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19783811

RESUMO

The farnesoid X receptor (FXR) is a bile acid activated nuclear receptor. Zucker (fa/fa) rats, harboring a loss of function mutation of the leptin receptor, develop diabetes, insulin resistance, obesity, and liver steatosis. In this study, we investigated the effect of FXR activation by 6-ethyl-chenodeoxycholic acid, (6E-CDCA, 10 mg/kg) on insulin resistance and liver and muscle lipid metabolism in fa/fa rats and compared its activity with rosiglitazone (10 mg/kg) alone or in combination with 6E-CDCA (5 mg/kg each). In comparison to lean (fa/+), fa/fa rats on a normal diet developed insulin resistance and liver steatosis. FXR activation protected against body weight gain and liver and muscle fat deposition and reversed insulin resistance as assessed by insulin responsive substrate-1 phosphorylation on serine 312 in liver and muscles. Activation of FXR reduced liver expression of genes involved in fatty acid synthesis, lipogenesis, and gluconeogenesis. In the muscles, FXR treatment reduced free fatty acid synthesis. Rosiglitazone reduced blood insulin, glucose, triglyceride, free fatty acid, and cholesterol plasma levels but promoted body weight gain (20%) and liver fat deposition. FXR activation reduced high density lipoprotein plasma levels. In summary, FXR administration reversed insulin resistance and correct lipid metabolism abnormalities in an obesity animal model.


Assuntos
Ácido Quenodesoxicólico/análogos & derivados , Fígado Gorduroso/prevenção & controle , Hipolipemiantes/uso terapêutico , Resistência à Insulina , Metabolismo dos Lipídeos/efeitos dos fármacos , Obesidade/complicações , Receptores Citoplasmáticos e Nucleares/agonistas , Animais , Ácido Quenodesoxicólico/administração & dosagem , Ácido Quenodesoxicólico/uso terapêutico , Modelos Animais de Doenças , Quimioterapia Combinada , Fígado Gorduroso/sangue , Fígado Gorduroso/patologia , Regulação da Expressão Gênica/efeitos dos fármacos , Hipoglicemiantes/administração & dosagem , Hipoglicemiantes/uso terapêutico , Hipolipemiantes/administração & dosagem , Proteínas Substratos do Receptor de Insulina/metabolismo , Metabolismo dos Lipídeos/genética , Lipídeos/sangue , Fígado/efeitos dos fármacos , Fígado/metabolismo , Fígado/patologia , Masculino , Músculo Esquelético/efeitos dos fármacos , Músculo Esquelético/metabolismo , Obesidade/sangue , Fosforilação/efeitos dos fármacos , RNA Mensageiro/metabolismo , Distribuição Aleatória , Ratos , Ratos Zucker , Receptores Citoplasmáticos e Nucleares/genética , Receptores Citoplasmáticos e Nucleares/metabolismo , Rosiglitazona , Tiazolidinedionas/administração & dosagem , Tiazolidinedionas/uso terapêutico , Fatores de Tempo
14.
J Cell Mol Med ; 14(1-2): 79-92, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-20041971

RESUMO

Atherosclerosis is the leading cause of illness and death. Therapeutic strategies aimed at reducing cholesterol plasma levels have shown efficacy in either reducing progression of atherosclerotic plaques and atherosclerosis-related mortality. The farnesoid-X-receptor (FXR) is a member of metabolic nuclear receptors (NRs) superfamily activated by bile acids. In entero-hepatic tissues, FXR functions as a bile acid sensor regulating bile acid synthesis, detoxification and excretion. In the liver FXR induces the expression of an atypical NR, the small heterodimer partner, which subsequently inhibits the activity of hepatocyte nuclear factor 4alpha repressing the transcription of cholesterol 7a-hydroxylase, the critical regulatory gene in bile acid synthesis. In the intestine FXR induces the release of fibroblast growth factor 15 (FGF15) (or FGF19 in human), which activates hepatic FGF receptor 4 (FGFR4) signalling to inhibit bile acid synthesis. In rodents, FXR activation decreases bile acid synthesis and lipogenesis and increases lipoprotein clearance, and regulates glucose homeostasis by reducing liver gluconeogenesis. FXR exerts counter-regulatory effects on macrophages, vascular smooth muscle cells and endothelial cells. FXR deficiency in mice results in a pro-atherogenetic lipoproteins profile and insulin resistance but FXR(-/-) mice fail to develop any detectable plaques on high-fat diet. Synthetic FXR agonists protect against development of aortic plaques formation in murine models characterized by pro-atherogenetic lipoprotein profile and accelerated atherosclerosis, but reduce HDL levels. Because human and mouse lipoprotein metabolism is modulated by different regulatory pathways the potential drawbacks of FXR ligands on HDL and bile acid synthesis need to addressed in relevant clinical settings.


Assuntos
Aterosclerose/tratamento farmacológico , Aterosclerose/metabolismo , Aterosclerose/fisiopatologia , Receptores Citoplasmáticos e Nucleares/metabolismo , Animais , Aterosclerose/patologia , Ácidos e Sais Biliares/metabolismo , Colesterol/metabolismo , Glucose/metabolismo , Humanos , Insulina/metabolismo , Fígado/citologia , Fígado/metabolismo , Modelos Moleculares , Estrutura Terciária de Proteína , Receptores Citoplasmáticos e Nucleares/química , Receptores Citoplasmáticos e Nucleares/genética , Transdução de Sinais/fisiologia , Triglicerídeos/metabolismo
15.
Biochim Biophys Acta ; 1792(6): 564-73, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19393742

RESUMO

Nuclear receptors are a family of ligand regulated factors that exert homeostatic functions at the interface between metabolic and immune function. The farnesoid X receptor (FXR) is a bile acid sensor expressed in immune cells such as macrophages where it exerts counter-regulatory effects. FXR deficient mice demonstrate disregulated immune response. Expression of FXR is down-regulated in inflamed tissues but the mechanism that leads to FXR down-regulation by inflammatory mediators is unknown. In the present study we have investigated the effect of inflammation-related cytokines on macrophages and demonstrated that INFgamma is a potent inhibitor of FXR gene expression/function in macrophages. STAT1 silencing and over-expression experiments demonstrated that FXR repression is mediated by INFgamma dependent activation of STAT1. Since IFNgamma is a potent activator of STAT1 we searched for STAT1 binding sites in the human FXR genomic and identified a region of the human FXR gene between the second and third exon that contains three hypothetical STAT1 binding sites. RAW 264.7 transiently transfected with an FXR genomic reporter construct which contained the three STAT binding sites responded to IFNgamma with a robust decrease in the reporter activity, demonstrating the potent modulation of FXR transcription by IFNgamma. Chromatin immunoprecipitation assay revealed that this region was immunoprecipitated following treatment of macrophage cell lines and supershift assay demonstrated that STAT1 was able to bind one of three identified sites. In summary, these results suggest that IFNgamma induced STAT1 homodimers modulate the transcriptional repression of FXR gene in macrophages during inflammation-related cytokines.


Assuntos
Inflamação/metabolismo , Interferon gama/farmacologia , Macrófagos/metabolismo , Receptores Citoplasmáticos e Nucleares/metabolismo , Fator de Transcrição STAT1/metabolismo , Sítios de Ligação , Linhagem Celular , Regulação da Expressão Gênica , Humanos , Interferon gama/metabolismo , Ativação de Macrófagos , Receptores Citoplasmáticos e Nucleares/antagonistas & inibidores , Receptores Citoplasmáticos e Nucleares/genética , Sequências Reguladoras de Ácido Nucleico , Fator de Transcrição STAT1/genética , Transdução de Sinais , Transfecção
16.
Mol Pain ; 6: 36, 2010 Jun 11.
Artigo em Inglês | MEDLINE | ID: mdl-20540729

RESUMO

BACKGROUND: Hydrogen sulphide (H2S) is a gaseous neuro-mediator that exerts analgesic effects in rodent models of visceral pain by activating KATP channels. A body of evidence support the notion that KATP channels interact with endogenous opioids. Whether H2S-induced analgesia involves opioid receptors is unknown. METHODS: The perception of painful sensation induced by colorectal distension (CRD) in conscious rats was measured by assessing the abdominal withdrawal reflex. The contribution of opioid receptors to H2S-induced analgesia was investigated by administering rats with selective mu, kappa and delta opioid receptor antagonists and antisenses. To investigate whether H2S causes mu opioid receptor (MOR) transactivation, the neuronal like cells SKNMCs were challenged with H2S in the presence of MOR agonist (DAMGO) or antagonist (CTAP). MOR activation and phosphorylation, its association to beta arrestin and internalization were measured. RESULTS: H2S exerted a potent analgesic effects on CRD-induced pain. H2S-induced analgesia required the activation of the opioid system. By pharmacological and molecular analyses, a robust inhibition of H2S-induced analgesia was observed in response to central administration of CTAP and MOR antisense, while kappa and delta receptors were less involved. H2S caused MOR transactivation and internalization in SKNMCs by a mechanism that required AKT phosphorylation. MOR transactivation was inhibited by LY294002, a PI3K inhibitor, and glibenclamide, a KATP channels blocker. CONCLUSIONS: This study provides pharmacological and molecular evidence that antinociception exerted by H2S in a rodent model of visceral pain is modulated by the transactivation of MOR. This observation provides support for development of new pharmacological approaches to visceral pain.


Assuntos
Analgesia , Analgésicos/metabolismo , Sulfeto de Hidrogênio/metabolismo , Dor/metabolismo , Receptores Opioides mu/metabolismo , Sulfetos/farmacologia , Animais , Linhagem Celular , Modelos Animais de Doenças , Humanos , Canais KATP/agonistas , Canais KATP/antagonistas & inibidores , Canais KATP/metabolismo , Masculino , Fosfatidilinositol 3-Quinases/metabolismo , Canais de Potássio/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Ratos , Ratos Wistar , Receptores Opioides mu/antagonistas & inibidores , Vísceras
17.
J Leukoc Biol ; 107(5): 797-807, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-31922289

RESUMO

Recently a G-protein-coupled receptor, MAS Related GPR Family Member X2 (MRGPRX2), was identified as a specific receptor on human mast cells responsible for IgE independent adverse drug reactions (ADR). Although a murine homologue, Mrgprb2, has been identified for this receptor, its affinity for many ADR-causing drugs is poor making it difficult to undertake in vivo studies to examine mechanisms of ADR and to develop therapeutic strategies. Here, we have created humanized mice capable of generating MRGPRX2-expressing human MCs allowing for the study of MRGPRX2 MCs-mediated ADR in vitro as well as in vivo. Humanized mice were generated by hydrodynamic-injection of plasmids expressing human GM-CSF and IL-3 into NOD-scid IL2R-γ-/- strain of mice that had been transplanted with human hematopoietic stem cells. These GM/IL-3 humice expressed high numbers of tissue human MCs but the MRGPRX2 receptor expressed in MCs were limited to few body sites including the skin. Importantly, large numbers of MRGPRX2-expressing human MCs could be cultured from the bone marrow of GM/IL-3 humice revealing these mice to be an important source of human MCs for in vitro studies of MRGPRX2-related MCs activities. When GM/IL-3 humice were exposed to known ADR causing contrast agents (meglumine and gadobutrol), the humice were found to experience anaphylaxis analogous to the clinical situation. Thus, GM/IL-3 humice represent a valuable model for investigating in vivo interactions of ADR-causing drugs and human MCs and their sequelae, and these mice are also a source of human MRGPRX2-expressing MCs for in vitro studies.


Assuntos
Modelos Animais de Doenças , Toxidermias/imunologia , Mastócitos/imunologia , Proteínas do Tecido Nervoso/imunologia , Receptores Acoplados a Proteínas G/imunologia , Receptores de Neuropeptídeos/imunologia , Animais , Meios de Contraste/toxicidade , Fator Estimulador de Colônias de Granulócitos e Macrófagos/genética , Humanos , Interleucina-3/genética , Mastócitos/efeitos dos fármacos , Meglumina/toxicidade , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Camundongos Transgênicos , Compostos Organometálicos/toxicidade
18.
Hepatology ; 47(2): 659-67, 2008 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-18098324

RESUMO

UNLABELLED: Increased intrahepatic resistance in cirrhotic livers is caused by endothelial dysfunction and impaired formation of two gaseous vasodilators, nitric oxide (NO) and hydrogen sulfide (H(2)S). Homocysteine, a sulfur-containing amino acid and H(2)S precursor, is formed from hepatic methionine metabolism. In the systemic circulation, hyperhomocystenemia impairs vasodilation and NO production from endothelial cells. Increased blood levels of homocysteine are common in patients with liver cirrhosis. In this study, we demonstrate that acute liver perfusion with homocysteine impairs NO formation and intrahepatic vascular relaxation induced by acetylcholine in methoxamine-precontracted normal livers (7.3% +/- 3.0% versus 26% +/- 2.7%; P < 0.0001). In rats with mild, diet-induced hyperhomocystenemia, the vasodilating activity of acetylcholine was markedly attenuated, and incremental increases in flow induced a greater percentage of increases in perfusion pressure than in control livers. Compared with normal rats, animals rendered cirrhotic by 12 weeks' administration of carbon tetrachloride exhibited a greater percentage of increments in perfusion pressure in response to shear stress (P < 0.05), and intrahepatic resistance to incremental increases in flow was further enhanced by homocysteine (P < 0.05). In normal hyperhomocysteinemic and cirrhotic rat livers, endothelial dysfunction caused by homocysteine was reversed by perfusion of the livers with sodium sulfide. Homocysteine reduced NO release from sinusoidal endothelial cells and also caused hepatic stellate cell contraction; this suggests a dual mechanism of action, with the latter effect being counteracted by H(2)S. CONCLUSION: Impaired vasodilation and hepatic stellate cell contraction caused by homocysteine contribute to the dynamic component of portal hypertension.


Assuntos
Homocisteína/farmacologia , Sulfeto de Hidrogênio/farmacologia , Circulação Hepática/fisiologia , Fígado/fisiologia , Metionina/fisiologia , Microcirculação/fisiologia , Acetilcolina/farmacologia , Animais , Modelos Animais de Doenças , Hiper-Homocisteinemia/sangue , Hiper-Homocisteinemia/fisiopatologia , Fígado/efeitos dos fármacos , Circulação Hepática/efeitos dos fármacos , Cirrose Hepática/sangue , Cirrose Hepática/fisiopatologia , Masculino , Microcirculação/efeitos dos fármacos , Ratos , Ratos Wistar
19.
Pharmacol Res ; 59(5): 319-29, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-19416632

RESUMO

Irritable bowel syndrome (IBS) is characterized by dysfunction of the afferent pathways that may lead to visceral hypersensitivity. Trimebutine is a weak opioid receptor agonist used in the treatment of IBS. We report on the effects of a novel derivative in which trimebutine has been salified with nitro-arginine(NO2-Arg-Trim), in modulating nociception to colorectal distension (CRD) in intact and post-colitis rats,an animal model that mimics some features of IBS. Colorectal sensitivity and pain were assessed by measuring the abdominal withdrawal score (AWR) during CRD. Healthy rats were treated with vehicle,trimebutine (10 mg/kg i.p.) or NO2-Arg-Trim (4, 8 and 16 mg/kg i.p.). Post-colitis, allodynic rats were investigated 4 weeks after colitis induction. Treating healthy rats with NO2-Arg-Trim resulted in a dose-dependent attenuation of CRD-induced nociception and in an inhibition of CRD-induced overexpression of spinal cFOS mRNA. NO2-Arg-Trim-induced antinociception was reversed by the opioid receptor antagonist naloxone and by the NO synthase-cGMP pathway inhibitor methylene blue, while L-NAME had no effect.The antinociceptive effect of NO2-Arg-Trim was maintained in a rodent model of post-inflammatory allodynia. In this setting,NO2-Arg-Trim but not trimebutine, significantly down-regulated the spinal cFOS mRNA expression and increased blood concentrations of NO2 +NO3. Moreover, the expression of several genes involved in inflammation and pain, as IL-1beta, TNFalpha, COX2 and iNOS, was up-regulated in colonic tissue from post-colitis rats and NO2-Arg-Trim, but not trimebutine, effectively reversed this effect. In summary, these data suggest that NO2-Arg-Trim inhibits nociception induced by CRD in both healthy and post-colitis, allodynic rats. The NO2-arginine moiety interacts with the opioid agonist trimebutine to potentiate its analgesic activity. This study provides evidence that NO2-arginine derivative of trimebutine might have beneficial effect in the treatment of painful intestinal disorders.


Assuntos
Colo/efeitos dos fármacos , Fármacos Gastrointestinais/farmacologia , Dor/tratamento farmacológico , Reto/efeitos dos fármacos , Trimebutina/análogos & derivados , Animais , Colite/fisiopatologia , Colo/metabolismo , Colo/fisiopatologia , Dilatação Patológica/fisiopatologia , Fármacos Gastrointestinais/uso terapêutico , Perfilação da Expressão Gênica , Inflamação/metabolismo , Inflamação/fisiopatologia , Masculino , Óxido Nítrico/fisiologia , Dor/metabolismo , Dor/fisiopatologia , Medição da Dor , Ratos , Ratos Wistar , Receptores Opioides mu/agonistas , Receptores Opioides mu/antagonistas & inibidores , Reto/fisiopatologia , Medula Espinal/metabolismo , Trimebutina/farmacologia , Trimebutina/uso terapêutico
20.
Front Immunol ; 10: 1566, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31379813

RESUMO

The inflammasome is a multi-protein complex that mediates proteolytic cleavage and release of the pro-inflammatory cytokines IL-1ß and IL-18, and pyroptosis-a form of cell death induced by various pathogenic bacteria. Apoptosis-associated speck-like protein containing a CARD (ASC) has a pivotal role in inflammasome assembly and activation. While ASC function has been primarily implicated in innate immune cells, its contribution to lymphocyte biology is unclear. Here we report that ASC is constitutively expressed in naïve CD4+ T cells together with the inflammasome sensor NLRP3 and caspase-1. When adoptively transferred in immunocompromised Rag1-/- mice, Asc-/- CD4+ T cells exacerbate T-cell-mediated autoimmune colitis. Asc-/- CD4+ T cells exhibit a higher proliferative capacity in vitro than wild-type CD4+ T cells. The increased expansion of Asc-/- CD4+ T cells in vivo correlated with robust TCR-mediated activation, inflammatory activity, and higher metabolic profile toward a highly glycolytic phenotype. These findings identify ASC as a crucial intrinsic regulator of CD4+ T-cell expansion that serves to maintain intestinal homeostasis.


Assuntos
Proteínas Adaptadoras de Sinalização CARD/imunologia , Linfócitos T CD4-Positivos/imunologia , Proliferação de Células , Homeostase/imunologia , Intestinos/imunologia , Animais , Apoptose/genética , Apoptose/imunologia , Proteínas Adaptadoras de Sinalização CARD/genética , Proteínas Adaptadoras de Sinalização CARD/metabolismo , Linfócitos T CD4-Positivos/citologia , Linfócitos T CD4-Positivos/metabolismo , Caspase 1/genética , Caspase 1/imunologia , Caspase 1/metabolismo , Células Cultivadas , Colite/genética , Colite/imunologia , Colite/metabolismo , Homeostase/genética , Inflamassomos/genética , Inflamassomos/imunologia , Ativação Linfocitária/genética , Ativação Linfocitária/imunologia , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos , Camundongos Knockout , Proteína 3 que Contém Domínio de Pirina da Família NLR/genética , Proteína 3 que Contém Domínio de Pirina da Família NLR/imunologia , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA