Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Immunol ; 202(5): 1510-1520, 2019 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-30683698

RESUMO

Macrophages exist as innate immune subsets that exhibit phenotypic heterogeneity and functional plasticity. Their phenotypes are dictated by inputs from the tissue microenvironment. G-protein-coupled receptors are essential in transducing signals from the microenvironment, and heterotrimeric Gα signaling links these receptors to downstream effectors. Several Gαi-coupled G-protein-coupled receptors have been implicated in macrophage polarization. In this study, we use genetically modified mice to investigate the role of Gαi2 on inflammasome activity and macrophage polarization. We report that Gαi2 in murine bone marrow-derived macrophages (BMDMs) regulates IL-1ß release after activation of the NLRP3, AIM2, and NLRC4 inflammasomes. We show this regulation stems from the biased polarity of Gαi2 deficient (Gnai2 -/-) and RGS-insensitive Gαi2 (Gnai2 G184S/G184S) BMDMs. We determined that although Gnai2 G184S/G184S BMDMs (excess Gαi2 signaling) have a tendency toward classically activated proinflammatory (M1) phenotype, Gnai2-/- BMDMs (Gαi2 deficient) are biased toward alternatively activated anti-inflammatory (M2) phenotype. Finally, we find that Gαi2-deficient macrophages have increased Akt activation and IFN-ß production but defects in ERK1/2 and STAT3 activation after LPS stimulation. Gαi2-deficient macrophages also exhibit increased STAT6 activation after IL-4 stimulation. In summary, our data indicates that excess Gαi2 signaling promotes an M1 macrophage phenotype, whereas Gαi2 signaling deficiency promotes an M2 phenotype. Understanding Gαi2-mediated effects on macrophage polarization may bring to light insights regarding disease pathogenesis and the reprogramming of macrophages for the development of novel therapeutics.


Assuntos
Citocinas/biossíntese , Subunidade alfa Gi2 de Proteína de Ligação ao GTP/imunologia , Inflamassomos/imunologia , Macrófagos/imunologia , Transdução de Sinais/imunologia , Animais , Células Cultivadas , Subunidade alfa Gi2 de Proteína de Ligação ao GTP/deficiência , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fenótipo
2.
Am J Physiol Cell Physiol ; 314(5): C616-C626, 2018 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-29342363

RESUMO

G protein-gated inwardly rectifying K+ (GIRK) channels are the major inwardly rectifying K+ currents in cardiac atrial myocytes and an important determinant of atrial electrophysiology. Inhibitory G protein α-subunits can both mediate activation via acetylcholine but can also suppress basal currents in the absence of agonist. We studied this phenomenon using whole cell patch clamping in murine atria from mice with global genetic deletion of Gαi2, combined deletion of Gαi1/Gαi3, and littermate controls. We found that mice with deletion of Gαi2 had increased basal and agonist-activated currents, particularly in the right atria while in contrast those with Gαi1/Gαi3 deletion had reduced currents. Mice with global genetic deletion of Gαi2 had decreased action potential duration. Tissue preparations of the left atria studied with a multielectrode array from Gαi2 knockout mice showed a shorter effective refractory period, with no change in conduction velocity, than littermate controls. Transcriptional studies revealed increased expression of GIRK channel subunit genes in Gαi2 knockout mice. Thus different G protein isoforms have differential effects on GIRK channel behavior and paradoxically Gαi2 act to increase basal and agonist-activated GIRK currents. Deletion of Gαi2 is potentially proarrhythmic in the atria.


Assuntos
Canais de Potássio Corretores do Fluxo de Internalização Acoplados a Proteínas G/metabolismo , Subunidades alfa de Proteínas de Ligação ao GTP/metabolismo , Átrios do Coração/metabolismo , Ativação do Canal Iônico , Potássio/metabolismo , Potenciais de Ação , Animais , Arritmias Cardíacas/genética , Arritmias Cardíacas/metabolismo , Arritmias Cardíacas/fisiopatologia , Função do Átrio Esquerdo , Função do Átrio Direito , Feminino , Subunidade alfa Gi2 de Proteína de Ligação ao GTP/deficiência , Subunidade alfa Gi2 de Proteína de Ligação ao GTP/metabolismo , Subunidades alfa de Proteínas de Ligação ao GTP/deficiência , Subunidades alfa de Proteínas de Ligação ao GTP/genética , Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/genética , Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/metabolismo , Frequência Cardíaca , Cinética , Masculino , Camundongos da Linhagem 129 , Camundongos Knockout , Período Refratário Eletrofisiológico
3.
J Neurochem ; 140(1): 82-95, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-27787898

RESUMO

We analyzed the role of a heterotrimeric G-protein, Gi2, in the development of the cerebral cortex. Acute knockdown of the α-subunit (Gαi2) with in utero electroporation caused delayed radial migration of excitatory neurons during corticogenesis, perhaps because of impaired morphology. The migration phenotype was rescued by an RNAi-resistant version of Gαi2. On the other hand, silencing of Gαi2 did not affect axon elongation, dendritic arbor formation or neurogenesis at ventricular zone in vivo. When behavior analyses were conducted with acute Gαi2-knockdown mice, they showed defects in social interaction, novelty recognition and active avoidance learning as well as increased anxiety. Subsequently, using whole-exome sequencing analysis, we identified a de novo heterozygous missense mutation (c.680C>T; p.Ala227Val) in the GNAI2 gene encoding Gαi2 in an individual with periventricular nodular heterotopia and intellectual disability. Collectively, the phenotypes in the knockdown experiments suggest a role of Gαi2 in the brain development, and impairment of its function might cause defects in neuronal functions which lead to neurodevelopmental disorders.


Assuntos
Córtex Cerebral/crescimento & desenvolvimento , Córtex Cerebral/metabolismo , Subunidade alfa Gi2 de Proteína de Ligação ao GTP/fisiologia , Deficiência Intelectual/metabolismo , Heterotopia Nodular Periventricular/metabolismo , Animais , Aprendizagem da Esquiva/fisiologia , Células COS , Córtex Cerebral/diagnóstico por imagem , Chlorocebus aethiops , Feminino , Subunidade alfa Gi2 de Proteína de Ligação ao GTP/deficiência , Humanos , Deficiência Intelectual/diagnóstico por imagem , Deficiência Intelectual/genética , Camundongos , Camundongos Endogâmicos ICR , Camundongos Knockout , Heterotopia Nodular Periventricular/diagnóstico por imagem , Heterotopia Nodular Periventricular/genética , Gravidez
4.
J Am Heart Assoc ; 5(11)2016 10 26.
Artigo em Inglês | MEDLINE | ID: mdl-27912212

RESUMO

BACKGROUND: We previously showed that the levels of both Giα-2 and Giα-3 proteins were augmented in spontaneously hypertensive rats (SHRs) before the onset of hypertension. In addition, intraperitoneal injection of pertussis toxin, which inactivates both Giα proteins, prevented the development of hypertension in SHRs. The aim of the present study was to determine the specific contributions of Giα-2 and Giα-3 proteins to the development of hypertension. METHODS AND RESULTS: Antisense oligodeoxynucleotide of Giα-2 and Giα-3 encapsulated in PEG/DOTAP/DOPE cationic liposomes were administrated intravenously into 3-week-old prehypertensive SHRs and Wistar Kyoto rats, whereas the control Wistar Kyoto rats and SHRs received PBS, empty liposomes, or sense. The knockdown of Giα-2 but not Giα-3 protein attenuated tachycardia and prevented the development of hypertension up to age 6 weeks; thereafter, blood pressure started increasing and reached the same level as that of untreated SHRs at 9 weeks. Furthermore, Giα-2 and Giα-3 antisense oligodeoxynucleotide treatments significantly decreased the enhanced levels of Giα-2 and Giα-3 proteins, respectively, and enhanced levels of superoxide anion and NADPH oxidase activity in heart, aorta, and kidney and hyperproliferation of vascular smooth muscle cells from SHRs aged 6 weeks. In addition, antisense oligodeoxynucleotide treatment with Giα-2 but not Giα-3 restored enhanced inhibition of adenylyl cyclase by oxotremorine to WKY levels. CONCLUSIONS: These results suggested that the enhanced expression of Giα-2 but not Giα-3 protein plays an important role in the pathogenesis of hypertension and tachycardia in SHRs.


Assuntos
Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/fisiologia , Hipertensão/prevenção & controle , Oligodesoxirribonucleotídeos Antissenso/fisiologia , Taquicardia/prevenção & controle , Inibidores de Adenilil Ciclases/farmacologia , Animais , Aorta/metabolismo , Pressão Sanguínea/fisiologia , Células Cultivadas , Modelos Animais de Doenças , Subunidade alfa Gi2 de Proteína de Ligação ao GTP/deficiência , Subunidade alfa Gi2 de Proteína de Ligação ao GTP/fisiologia , Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/deficiência , Técnicas de Silenciamento de Genes , Frequência Cardíaca/fisiologia , Rim/metabolismo , Lipossomos/administração & dosagem , Masculino , Músculo Liso Vascular/metabolismo , Miocárdio/metabolismo , Ratos Endogâmicos SHR , Ratos Endogâmicos WKY , Transdução de Sinais/fisiologia , Transfecção/métodos
5.
FEBS Lett ; 590(23): 4361-4371, 2016 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-27718249

RESUMO

Despite the importance of glutathione in Dictyostelium, the role of glutathione synthetase (gshB/GSS) has not been clearly investigated. In this study, we observed that increasing glutathione content by constitutive expression of gshB leads to mound-arrest and defects in 3',5'-cyclic adenosine monophosphate (cAMP)-mediated aggregation and developmental gene expression. The overexpression of gpaB encoding G protein alpha 2 (Gα2), an essential component of the cAMP signalling pathway, results in a phenotype similar to that caused by gshB overexpression, whereas gpaB knockdown in gshB-overexpressing cells partially rescues the above-mentioned phenotypic defects. Furthermore, Gα2 is highly enriched at the plasma membrane of gshB-overexpressing cells compared to wild-type cells. Therefore, our findings suggest that glutathione upregulates cAMP signalling via Gα2 modulation during Dictyostelium development.


Assuntos
AMP Cíclico/metabolismo , Dictyostelium/citologia , Dictyostelium/crescimento & desenvolvimento , Subunidade alfa Gi2 de Proteína de Ligação ao GTP/metabolismo , Glutationa/metabolismo , Regulação para Cima , Membrana Celular/metabolismo , Dictyostelium/genética , Dictyostelium/metabolismo , Subunidade alfa Gi2 de Proteína de Ligação ao GTP/deficiência , Subunidade alfa Gi2 de Proteína de Ligação ao GTP/genética , Técnicas de Silenciamento de Genes , Glutationa Sintase/genética , Espaço Intracelular/metabolismo , Transdução de Sinais
6.
Sci Rep ; 6: 30925, 2016 08 08.
Artigo em Inglês | MEDLINE | ID: mdl-27499046

RESUMO

Putative functions of the heterotrimeric G-protein subunit Gαi2-dependent signaling include ion channel regulation, cell differentiation, proliferation and apoptosis. Erythrocytes may, similar to apoptosis of nucleated cells, undergo eryptosis, characterized by cell shrinkage and cell membrane scrambling with phosphatidylserine (PS) exposure. Eryptosis may be triggered by increased cytosolic Ca(2+) activity and ceramide. In the present study, we show that Gαi2 is expressed in both murine and human erythrocytes and further examined the survival of erythrocytes drawn from Gαi2-deficient mice (Gαi2(-/-)) and corresponding wild-type mice (Gαi2(+/+)). Our data show that plasma erythropoietin levels, erythrocyte maturation markers, erythrocyte counts, hematocrit and hemoglobin concentration were similar in Gαi2(-/-) and Gαi2(+/+) mice but the mean corpuscular volume was significantly larger in Gαi2(-/-) mice. Spontaneous PS exposure of circulating Gαi2(-/-) erythrocytes was significantly lower than that of circulating Gαi2(+/+) erythrocytes. PS exposure was significantly lower in Gαi2(-/-) than in Gαi2(+/+) erythrocytes following ex vivo exposure to hyperosmotic shock, bacterial sphingomyelinase or C6 ceramide. Erythrocyte Gαi2 deficiency further attenuated hyperosmotic shock-induced increase of cytosolic Ca(2+) activity and cell shrinkage. Moreover, Gαi2(-/-) erythrocytes were more resistant to osmosensitive hemolysis as compared to Gαi2(+/+) erythrocytes. In conclusion, Gαi2 deficiency in erythrocytes confers partial protection against suicidal cell death.


Assuntos
Eriptose , Eritrócitos/citologia , Eritrócitos/fisiologia , Subunidade alfa Gi2 de Proteína de Ligação ao GTP/metabolismo , Animais , Sobrevivência Celular , Índices de Eritrócitos , Eritrócitos/química , Subunidade alfa Gi2 de Proteína de Ligação ao GTP/deficiência , Humanos , Camundongos , Camundongos Knockout , Fosfatidilserinas/análise
7.
Cardiovasc Res ; 108(3): 348-56, 2015 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-26464333

RESUMO

AIMS: Inhibitory G (Gi) proteins have been proposed to be cardioprotective. We investigated effects of Gαi2 knockout on cardiac function and survival in a murine heart failure model of cardiac ß1-adrenoceptor overexpression. METHODS AND RESULTS: ß1-transgenic mice lacking Gαi2 (ß1-tg/Gαi2 (-/-)) were compared with wild-type mice and littermates either overexpressing cardiac ß1-adrenoceptors (ß1-tg) or lacking Gαi2 (Gαi2 (-/-)). At 300 days, mortality of mice only lacking Gαi2 was already higher compared with wild-type or ß1-tg, but similar to ß1-tg/Gαi2 (-/-), mice. Beyond 300 days, mortality of ß1-tg/Gαi2 (-/-) mice was enhanced compared with all other genotypes (mean survival time: 363 ± 21 days). At 300 days of age, echocardiography revealed similar cardiac function of wild-type, ß1-tg, and Gαi2 (-/-) mice, but significant impairment for ß1-tg/Gαi2 (-/-) mice (e.g. ejection fraction 14 ± 2 vs. 40 ± 4% in wild-type mice). Significantly increased ventricle-to-body weight ratio (0.71 ± 0.06 vs. 0.48 ± 0.02% in wild-type mice), left ventricular size (length 0.82 ± 0.04 vs. 0.66 ± 0.03 cm in wild types), and atrial natriuretic peptide and brain natriuretic peptide expression (mRNA: 2819 and 495% of wild-type mice, respectively) indicated hypertrophy. Gαi3 was significantly up-regulated in Gαi2 knockout mice (protein compared with wild type: 340 ± 90% in Gαi2 (-/-) and 394 ± 80% in ß1-tg/Gαi2 (-/-), respectively). CONCLUSIONS: Gαi2 deficiency combined with cardiac ß1-adrenoceptor overexpression strongly impaired survival and cardiac function. At 300 days of age, ß1-adrenoceptor overexpression alone had not induced cardiac hypertrophy or dysfunction while there was overt cardiomyopathy in mice additionally lacking Gαi2. We propose an enhanced effect of increased ß1-adrenergic drive by the lack of protection via Gαi2. Gαi3 up-regulation was not sufficient to compensate for Gαi2 deficiency, suggesting an isoform-specific or a concentration-dependent mechanism.


Assuntos
Cardiomiopatia Dilatada/metabolismo , Subunidade alfa Gi2 de Proteína de Ligação ao GTP/deficiência , Insuficiência Cardíaca/metabolismo , Miócitos Cardíacos/metabolismo , Receptores Adrenérgicos beta 1/metabolismo , Animais , Fator Natriurético Atrial/genética , Fator Natriurético Atrial/metabolismo , Cardiomiopatia Dilatada/diagnóstico por imagem , Cardiomiopatia Dilatada/genética , Cardiomiopatia Dilatada/fisiopatologia , AMP Cíclico/metabolismo , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Modelos Animais de Doenças , Subunidade alfa Gi2 de Proteína de Ligação ao GTP/genética , Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/genética , Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/metabolismo , Regulação da Expressão Gênica , Predisposição Genética para Doença , Insuficiência Cardíaca/diagnóstico por imagem , Insuficiência Cardíaca/genética , Insuficiência Cardíaca/fisiopatologia , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Knockout , Miócitos Cardíacos/patologia , Peptídeo Natriurético Encefálico/genética , Peptídeo Natriurético Encefálico/metabolismo , Fenótipo , Receptores Adrenérgicos beta 1/genética , Volume Sistólico , Fatores de Tempo , Ultrassonografia , Função Ventricular Esquerda , Remodelação Ventricular
8.
Proc Natl Acad Sci U S A ; 112(20): 6491-6, 2015 May 19.
Artigo em Inglês | MEDLINE | ID: mdl-25944935

RESUMO

Platelets are crucial for hemostasis and thrombosis and exacerbate tissue injury following ischemia and reperfusion. Important regulators of platelet function are G proteins controlled by seven transmembrane receptors. The Gi protein Gα(i2) mediates platelet activation in vitro, but its in vivo role in hemostasis, arterial thrombosis, and postischemic infarct progression remains to be determined. Here we show that mice lacking Gα(i2) exhibit prolonged tail-bleeding times and markedly impaired thrombus formation and stability in different models of arterial thrombosis. We thus generated mice selectively lacking Gα(i2) in megakaryocytes and platelets (Gna(i2)(fl/fl)/PF4-Cre mice) and found bleeding defects comparable to those in global Gα(i2)-deficient mice. To examine the impact of platelet Gα(i2) in postischemic thrombo-inflammatory infarct progression, Gna(i2)(fl/fl)/PF4-Cre mice were subjected to experimental models of cerebral and myocardial ischemia/reperfusion injury. In the model of transient middle cerebral artery occlusion stroke Gna(i2)(fl/fl)/PF4-Cre mice developed significantly smaller brain infarcts and fewer neurological deficits than littermate controls. Following myocardial ischemia, Gna(i2)(fl/fl)/PF4-Cre mice showed dramatically reduced reperfusion injury which correlated with diminished formation of the ADP-dependent platelet neutrophil complex. In conclusion, our data provide definitive evidence that platelet Gα(i2) not only controls hemostatic and thrombotic responses but also is critical for the development of ischemia/reperfusion injury in vivo.


Assuntos
Subunidade alfa Gi2 de Proteína de Ligação ao GTP/metabolismo , Infarto da Artéria Cerebral Média/fisiopatologia , Inflamação/fisiopatologia , Ativação Plaquetária/fisiologia , Traumatismo por Reperfusão/fisiopatologia , Trombose/fisiopatologia , Animais , Tempo de Sangramento , Plaquetas/metabolismo , Subunidade alfa Gi2 de Proteína de Ligação ao GTP/deficiência , Immunoblotting , Megacariócitos/metabolismo , Camundongos , Traumatismo por Reperfusão/prevenção & controle
9.
Cell Signal ; 26(2): 409-18, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24511612

RESUMO

Cyclic AMP (cAMP) is an important intracellular signaling molecule for many G protein-mediated signaling pathways but the specificity of cAMP signaling in cells with multiple signaling pathways is not well-understood. In Dictyostelium, at least two different G protein signaling pathways, mediated by the Gα2 and Gα4 subunits, are involved with cAMP accumulation, spore production, and chemotaxis and the stimulation of these pathways results in the activation of ERK2, a mitogen-activated protein kinase that can down regulate the cAMP-specific phosphodiesterase RegA. The regA gene was disrupted in gα2(−) and gα4(−) cells to determine if the absence of this phosphodiesterase rescues the development of these G protein mutants as it does for erk2(−) mutants. There gA(−) mutation had no major effects on developmental morphology but enriched the distribution of the Gα mutant cells to the prespore/prestalk border in chimeric aggregates. The loss of RegA function had no effect on Gα4- mediated folate chemotaxis. However, the regA gene disruption in gα4(−) cells, but not in gα2(−) cells, resulted in a substantial rescue and acceleration of spore production. This rescue in sporulation required cell autonomous signaling because the precocious sporulation could not be induced through intercellular signaling in chimeric aggregates. However, intercellular signals from regA(−) strains increased the expression of the prestalk gene ecmB and accelerated the vacuolization of stalk cells. Intercellular signaling from the gα4(−)regA(−) strain did not induce ecmA gene expression indicating cell-type specificity in the promotion of prestalk cell development. regA gene disruption in a Gα4(HC) (Gα4 overexpression) strain did not result in precocious sporulation or stalk cell development indicating that elevated Gα4 subunit expression can mask regA(−) associated phenotypes even when provided with wild-type intercellular signaling. These findings indicate that the Gα2 and Gα4-mediated pathways provide different contributions to the development of spores and stalk cells and that the absence of RegA function can bypass some but not all defects in G protein regulated spore development.


Assuntos
3',5'-AMP Cíclico Fosfodiesterases/metabolismo , AMP Cíclico/metabolismo , Dictyostelium/enzimologia , Dictyostelium/genética , Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/metabolismo , Proteínas de Protozoários/metabolismo , 3',5'-AMP Cíclico Fosfodiesterases/genética , Dictyostelium/crescimento & desenvolvimento , Dictyostelium/metabolismo , Regulação para Baixo/efeitos dos fármacos , Proteínas da Matriz Extracelular/genética , Proteínas da Matriz Extracelular/metabolismo , Ácido Fólico/farmacologia , Subunidade alfa Gi2 de Proteína de Ligação ao GTP/deficiência , Subunidade alfa Gi2 de Proteína de Ligação ao GTP/genética , Subunidade alfa Gi2 de Proteína de Ligação ao GTP/metabolismo , Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/deficiência , Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/genética , Proteína Quinase 1 Ativada por Mitógeno/genética , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Mutação , Proteínas de Protozoários/genética , Transdução de Sinais , Esporos de Protozoários/enzimologia , Esporos de Protozoários/genética , Complexo Vitamínico B/farmacologia
10.
PLoS One ; 8(8): e72596, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23977324

RESUMO

B lymphocytes are compartmentalized within lymphoid organs. The organization of these compartments depends upon signaling initiated by G-protein linked chemoattractant receptors. To address the importance of the G-proteins Gαi2 and Gαi3 in chemoattractant signaling we created mice lacking both proteins in their B lymphocytes. While bone marrow B cell development and egress is grossly intact; mucosal sites, splenic marginal zones, and lymph nodes essentially lack B cells. There is a partial block in splenic follicular B cell development and a 50-60% reduction in splenic B cells, yet normal numbers of splenic T cells. The absence of Gαi2 and Gαi3 in B cells profoundly disturbs the architecture of lymphoid organs with loss of B cell compartments in the spleen, thymus, lymph nodes, and gastrointestinal tract. This results in a severe disruption of B cell function and a hyper-IgM like syndrome. Beyond the pro-B cell stage, B cells are refractory to chemokine stimulation, and splenic B cells are poorly responsive to antigen receptor engagement. Gαi2 and Gαi3 are therefore critical for B cell chemoattractant receptor signaling and for normal B cell function. These mice provide a worst case scenario of the consequences of losing chemoattractant receptor signaling in B cells.


Assuntos
Linfócitos B/metabolismo , Linfócitos B/patologia , Compartimento Celular , Subunidade alfa Gi2 de Proteína de Ligação ao GTP/deficiência , Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/deficiência , Síndrome de Imunodeficiência com Hiper-IgM/imunologia , Síndrome de Imunodeficiência com Hiper-IgM/patologia , Animais , Formação de Anticorpos/efeitos dos fármacos , Linfócitos B/efeitos dos fármacos , Medula Óssea/efeitos dos fármacos , Medula Óssea/patologia , Compartimento Celular/efeitos dos fármacos , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Quimiocinas/metabolismo , Fatores Quimiotáticos/farmacologia , Progressão da Doença , Subunidade alfa Gi2 de Proteína de Ligação ao GTP/metabolismo , Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/metabolismo , Centro Germinativo/efeitos dos fármacos , Centro Germinativo/patologia , Imunização , Integrases/metabolismo , Integrinas/metabolismo , Lipopolissacarídeos/farmacologia , Linfonodos/efeitos dos fármacos , Linfonodos/patologia , Contagem de Linfócitos , Subpopulações de Linfócitos/efeitos dos fármacos , Subpopulações de Linfócitos/metabolismo , Camundongos Endogâmicos C57BL , Camundongos Knockout , Peritônio/efeitos dos fármacos , Peritônio/patologia , Receptores de Antígenos de Linfócitos B/metabolismo , Transdução de Sinais/efeitos dos fármacos , Baço/patologia
11.
Am J Physiol Regul Integr Comp Physiol ; 305(4): R435-42, 2013 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-23697798

RESUMO

Reciprocal physiological modulation of heart rate is controlled by the sympathetic and parasympathetic systems acting on the sinoatrial (SA) node. However, there is little direct in vivo work examining the role of stimulatory and inhibitory G protein signaling in the SA node. Thus, we designed a study to examine the role of the stimulatory (Gαs) and inhibitory G protein (Gαi2) in in vivo heart rate regulation in the SA node in the mouse. We studied mice with conditional deletion of Gαs and Gαi2 in the conduction system using cre-loxP technology. We crossed mice in which cre recombinase expression was driven by a tamoxifen-inducible conduction system-specific construct with "Gαs floxed" and "Gαi2 floxed" mice. We studied the heart rate responses of adult mice compared with littermate controls by using radiotelemetry before and after administration of tamoxifen. The mice with conditional deletion of Gαs and Gαi2 had a loss of diurnal variation and were bradycardic or tachycardic, respectively, in the daytime. In mice with conditional deletion of Gαs, there was a selective loss of low-frequency power, while with deletion of Gαi2, there was a loss of high-frequency power in power spectral analysis of heart rate variability. There was no evidence of pathological arrhythmia. Pharmacological modulation of heart rate by isoprenaline was impaired in the Gαs mice, but a muscarinic agonist was still able to slow the heart rate in Gαi2 mice. We conclude that Gαs- and Gαi2-mediated signaling in the sinoatrial node is important in the reciprocal regulation of heart rate through the autonomic nervous system.


Assuntos
Subunidade alfa Gi2 de Proteína de Ligação ao GTP/metabolismo , Subunidades alfa Gs de Proteínas de Ligação ao GTP/metabolismo , Frequência Cardíaca , Nó Sinoatrial/metabolismo , Agonistas Adrenérgicos beta/farmacologia , Animais , Sistema Nervoso Autônomo/metabolismo , Sistema Nervoso Autônomo/fisiopatologia , Bradicardia/metabolismo , Bradicardia/fisiopatologia , Cromograninas , Ritmo Circadiano , Eletrocardiografia Ambulatorial/métodos , Subunidade alfa Gi2 de Proteína de Ligação ao GTP/deficiência , Subunidade alfa Gi2 de Proteína de Ligação ao GTP/genética , Subunidades alfa Gs de Proteínas de Ligação ao GTP/deficiência , Subunidades alfa Gs de Proteínas de Ligação ao GTP/genética , Frequência Cardíaca/efeitos dos fármacos , Integrases/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Agonistas Muscarínicos/farmacologia , Transdução de Sinais , Nó Sinoatrial/efeitos dos fármacos , Nó Sinoatrial/inervação , Nó Sinoatrial/fisiopatologia , Taquicardia/metabolismo , Taquicardia/fisiopatologia , Telemetria , Fatores de Tempo
12.
Inflamm Bowel Dis ; 19(4): 881-8, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23448795

RESUMO

Inflammatory bowel disease is characterized by a number of immunological alterations, not the least in the T-cell compartment. Numerous animal models of colitis have revealed aberrant thymocyte dynamics associated with skewed thymocyte development. The recent advancements in quantitative methods have proposed critical kinetic alterations in the thymocyte development during the progression of colitis. This review focuses on the aberrant thymocyte dynamics in Gαi2-deficient mice as this mouse model provides most quantitative data of the thymocyte development associated with colitis. Herein, we discuss several dynamic changes during the progression of colitis and propose a hypothesis for the underlying causes for the skewed proportions of the thymocyte populations seen in the Gαi2-deficient mice and in other mouse models of colitis.


Assuntos
Modelos Animais de Doenças , Subunidade alfa Gi2 de Proteína de Ligação ao GTP/deficiência , Doenças Inflamatórias Intestinais/etiologia , Doenças Inflamatórias Intestinais/patologia , Timócitos/patologia , Animais , Subunidade alfa Gi2 de Proteína de Ligação ao GTP/genética , Humanos , Camundongos
13.
Int Immunol ; 25(1): 35-44, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22962436

RESUMO

Gαi2-deficient mice spontaneously develop colitis. Using xMAP technology and RT-PCR, we investigated cytokine/chemokine profiles during histologically defined phases of disease: (i) no/mild, (ii) moderate, (iii) severe colitis without dysplasia/cancer and (iv) severe colitis with dysplasia/cancer, compared with age-matched wild-type (WT) littermates. Colonic dysplasia was observed in 4/11 mice and cancer in 1/11 mice with severe colitis. The histology correlated with progressive increases in colon weight/cm and spleen weight, and decreased thymus weight, all more advanced in mice with dysplasia/cancer. IL-1ß, IL-6, IL-12p40, IL-17, TNF-α, CCL2 and CXCL1 protein levels in colons, but not small intestines increased with colitis progression and were significantly increased in mice with moderate and severe colitis compared with WT mice, irrespective of the absence/presence of dysplasia/cancer. CCL5 did not change during colitis progression. Colonic IL-17 transcription increased 40- to 70-fold in all stages of colitis, whereas IFN-γ mRNA was gradually up-regulated 12- to 55-fold with colitis progression, and further to 62-fold in mice with dysplasia/cancer. IL-27 mRNA increased 4- to 15-fold during the course of colitis, and colonic IL-21 transcription increased 3-fold in mice with severe colitis, both irrespective of the absence/presence of dysplasia/cancer. FoxP3 transcription was significantly enhanced (3.5-fold) in mice with moderate and severe colitis, but not in mice with dysplasia/cancer, compared with WT mice. Constrained correspondence analysis demonstrated an association between increased protein levels of TNF-α, CCL2, IL-1ß, IL-6 and CXCL1 and dysplasia/cancer. In conclusion, colonic responses are dominated by a mixed T(h)1/T(h)17 phenotype, with increasing T(h)1 cytokine transcription with progression of colitis in Gαi2(-/-) mice.


Assuntos
Colite/imunologia , Colo/imunologia , Neoplasias do Colo/imunologia , Citocinas/imunologia , Subunidade alfa Gi2 de Proteína de Ligação ao GTP/deficiência , Células Th1/imunologia , Células Th17/imunologia , Animais , Colite/genética , Colite/metabolismo , Colite/patologia , Colo/patologia , Neoplasias do Colo/genética , Neoplasias do Colo/metabolismo , Neoplasias do Colo/patologia , Citocinas/genética , Feminino , Fatores de Transcrição Forkhead/genética , Fatores de Transcrição Forkhead/imunologia , Subunidade alfa Gi2 de Proteína de Ligação ao GTP/genética , Subunidade alfa Gi2 de Proteína de Ligação ao GTP/imunologia , Regulação da Expressão Gênica/imunologia , Intestino Delgado/imunologia , Intestino Delgado/patologia , Masculino , Camundongos , Camundongos Knockout , Tamanho do Órgão , Índice de Gravidade de Doença , Transdução de Sinais , Baço/imunologia , Baço/patologia , Células Th1/metabolismo , Células Th1/patologia , Equilíbrio Th1-Th2 , Células Th17/metabolismo , Células Th17/patologia , Timo/imunologia , Timo/patologia , Transcrição Gênica/imunologia
14.
J Immunol ; 190(1): 324-33, 2013 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-23225882

RESUMO

Heterotrimeric G proteins of the Gα(i) family have been implicated in signaling pathways regulating cell migration in immune diseases. The Gα(i)-protein-coupled C5a receptor is a critical regulator of IgG FcR function in experimental models of immune complex (IC)-induced inflammation. By using mice deficient for Gα(i2) or Gα(i3), we show that Gα(i2) is necessary for neutrophil influx in skin and lung Arthus reactions and agonist-induced neutrophilia in the peritoneum, whereas Gα(i3) plays a less critical but variable role. Detailed analyses of the pulmonary IC-induced inflammatory response revealed several shared functions of Gα(i2) and Gα(i3), including mediating C5a anaphylatoxin receptor-induced activation of macrophages, involvement in alveolar production of chemokines, transition of neutrophils from bone marrow into blood, and modulation of CD11b and CD62L expression that account for neutrophil adhesion to endothelial cells. Interestingly, C5a-stimulated endothelial polymorphonuclear neutrophil transmigration, but not chemotaxis, is enhanced versus reduced in the absence of neutrophil Gα(i3) or Gα(i2), respectively, and knockdown of endothelial Gα(i2) caused decreased transmigration of wild-type neutrophils. These data demonstrate that Gα(i2) and Gα(i3) contribute to inflammation by redundant, overlapping, and Gα(i)-isoform-specific mechanisms, with Gα(i2) exhibiting unique functions in both neutrophils and endothelial cells that appear essential for polymorphonuclear neutrophil recruitment in IC disease.


Assuntos
Lesão Pulmonar Aguda/imunologia , Subunidade alfa Gi2 de Proteína de Ligação ao GTP/fisiologia , Lesão Pulmonar Aguda/genética , Lesão Pulmonar Aguda/patologia , Animais , Reação de Arthus/genética , Reação de Arthus/imunologia , Reação de Arthus/patologia , Adesão Celular/genética , Adesão Celular/imunologia , Quimiotaxia de Leucócito/genética , Quimiotaxia de Leucócito/imunologia , Modelos Animais de Doenças , Regulação para Baixo/genética , Regulação para Baixo/imunologia , Endotélio Vascular/citologia , Endotélio Vascular/imunologia , Endotélio Vascular/patologia , Subunidade alfa Gi2 de Proteína de Ligação ao GTP/deficiência , Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/fisiologia , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neutrófilos/citologia , Neutrófilos/imunologia , Neutrófilos/patologia , Regulação para Cima/genética , Regulação para Cima/imunologia
15.
J Immunol ; 189(2): 980-7, 2012 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-22706085

RESUMO

Various heterotrimeric G(i) proteins are considered to be involved in cell migration and effector function of immune cells. The underlying mechanisms, how they control the activation of myeloid effector cells, are not well understood. To elucidate isoform-redundant and -specific roles for Gα(i) proteins in these processes, we analyzed mice genetically deficient in Gα(i2) or Gα(i3). First, we show an altered distribution of tissue macrophages and blood monocytes in the absence of Gα(i2) but not Gα(i3). Gα(i2)-deficient but not wild-type or Gα(i3)-deficient mice exhibited reduced recruitment of macrophages in experimental models of thioglycollate-induced peritonitis and LPS-triggered lung injury. In contrast, genetic ablation of Gα(i2) had no effect on Gα(i)-dependent peritoneal cytokine production in vitro and the phagocytosis-promoting function of the Gα(i)-coupled C5a anaphylatoxin receptor by liver macrophages in vivo. Interestingly, actin rearrangement and CCL2- and C5a anaphylatoxin receptor-induced chemotaxis but not macrophage CCR2 and C5a anaphylatoxin receptor expression were reduced in the specific absence of Gα(i2). Furthermore, knockdown of Gα(i2) caused decreased cell migration and motility of RAW 264.7 cells, which was rescued by transfection of Gα(i2) but not Gα(i3). These results indicate that Gα(i2), albeit redundant to Gα(i3) in some macrophage activation processes, clearly exhibits a Gα(i) isoform-specific role in the regulation of macrophage migration.


Assuntos
Inibição de Migração Celular/imunologia , Subunidade alfa Gi2 de Proteína de Ligação ao GTP/deficiência , Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/deficiência , Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/fisiologia , Macrófagos/imunologia , Macrófagos/patologia , Lesão Pulmonar Aguda/imunologia , Lesão Pulmonar Aguda/patologia , Animais , Inibição de Migração Celular/genética , Subunidade alfa Gi2 de Proteína de Ligação ao GTP/genética , Lipopolissacarídeos/toxicidade , Macrófagos/metabolismo , Camundongos , Camundongos da Linhagem 129 , Camundongos Knockout , Monócitos/imunologia , Monócitos/patologia , Peritonite/induzido quimicamente , Peritonite/imunologia , Peritonite/patologia , Tioglicolatos/toxicidade
16.
Am J Physiol Gastrointest Liver Physiol ; 300(6): G939-47, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21436315

RESUMO

The large (LI) and small intestine (SI) differ in patterns of susceptibility to chronic mucosal inflammation. In this study, we evaluated whether this might, in part, reflect differences in resident mucosal CD11c(+) T cells. These cells comprised 39-48% (SI) and 12-17% (LI) of the intraepithelial compartment, most of which were T-cell receptor-αß(+). In the SI, the majority of these cells were CD103(+) CD8(+) NK1.1(-), whereas the opposite phenotype prevailed in the LI. In transfer models of CD4(+) T cell-induced colitis, small numbers (2.5 × 10(5)) of SI CD11c(+) CD8(+) T cells suppressed proinflammatory cytokine-producing CD4(+) T cells in mesenteric lymph nodes and mucosa-associated lymphoid compartments (SI and LI) and protected mice from chronic inflammation. On a per-cell basis, the regulatory function of SI CD11c(+) T cells in CD4(+) T cell colitis was potent compared with other reported regulatory CD4(+) or CD8(+) T cells. In contrast, neither LI CD11c(+) T cells nor SI CD11c(-) T cells were effective in such immunoregulation. SI CD11c(+) CD8(+) T cells were similarly effective in suppressing CD4(+)CD45RB(hi) T cell colitis, as evidenced by inhibition of intracellular proinflammatory cytokine expression and histological inflammation. These findings indicate that SI CD11c(+) CD8(+) T cells are a distinct intestinal T cell population that plays an immunoregulatory role in control of proinflammatory CD4(+) T cells and maintenance of intestinal mucosal homeostasis.


Assuntos
Antígeno CD11c/metabolismo , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Colite/prevenção & controle , Íleo/imunologia , Jejuno/imunologia , Subpopulações de Linfócitos T/imunologia , Análise de Variância , Animais , Linfócitos T CD4-Positivos/transplante , Linfócitos T CD8-Positivos/transplante , Colite/imunologia , Colite/patologia , Colo/imunologia , Colo/patologia , Citocinas/metabolismo , Proteínas de Ligação a DNA/deficiência , Proteínas de Ligação a DNA/genética , Modelos Animais de Doenças , Subunidade alfa Gi2 de Proteína de Ligação ao GTP/deficiência , Subunidade alfa Gi2 de Proteína de Ligação ao GTP/genética , Genes Codificadores dos Receptores de Linfócitos T , Homeostase , Íleo/patologia , Imunidade nas Mucosas , Imunofenotipagem , Mediadores da Inflamação/metabolismo , Mucosa Intestinal/imunologia , Mucosa Intestinal/patologia , Jejuno/patologia , Antígenos Comuns de Leucócito/genética , Linfonodos/imunologia , Ativação Linfocitária , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos SCID , Camundongos Transgênicos , Fenótipo , Subpopulações de Linfócitos T/transplante , Fatores de Tempo
17.
Circ Arrhythm Electrophysiol ; 3(4): 391-400, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20495013

RESUMO

BACKGROUND: We explored the role that inhibitory heterotrimeric G-proteins play in ventricular arrhythmia. METHODS AND RESULTS: Mice with global genetic deletion of Galpha(i2) [Galpha(i2) (-/-)] were studied and found, based on telemetry, to have a prolonged QT interval on surface ECG when awake. In vivo electrophysiology studies revealed that the Galpha(i2) (-/-) mice have a reduced ventricular effective refractory period and a predisposition to ventricular tachycardia when challenged with programmed electrical stimulation. Neither control nor combined global deletion of Galpha(i1) and Galpha(i3) mice showed these abnormalities. There was no evidence for structural heart disease at this time point in the Galpha(i2) (-/-) mice as assessed by cardiac histology and echocardiography. The absence of Galpha(i2) thus leads to a primary electrical abnormality, and we explored the basis for this finding. With patch clamping, single isolated ventricular cells showed that Galpha(i2) (-/-) mice had a prolonged ventricular action potential duration (APD) but steeper action potential shortening as the diastolic interval was reduced in restitution studies. Gene expression studies showed increased expression of L-type Ca(2+) channel subunits, and patch clamping revealed an increase in these currents in Galpha(i2) (-/-) mice. There were no changes in K(+) currents. CONCLUSIONS: The absence of inhibitory G-protein signaling mediated through Galpha(i2) is a substrate for ventricular arrhythmias.


Assuntos
Subunidade alfa Gi2 de Proteína de Ligação ao GTP/deficiência , Sistema de Condução Cardíaco/metabolismo , Taquicardia Ventricular/metabolismo , Potenciais de Ação , Animais , Cálcio/metabolismo , Canais de Cálcio Tipo L/metabolismo , Estimulação Cardíaca Artificial , Modelos Animais de Doenças , Ecocardiografia Doppler de Pulso , Eletrocardiografia Ambulatorial , Subunidade alfa Gi2 de Proteína de Ligação ao GTP/genética , Sistema de Condução Cardíaco/fisiopatologia , Camundongos , Camundongos Knockout , Técnicas de Patch-Clamp , Potássio/metabolismo , Taquicardia Ventricular/genética , Taquicardia Ventricular/fisiopatologia , Telemetria , Fatores de Tempo
18.
J Immunol ; 179(1): 439-48, 2007 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-17579064

RESUMO

Signals generated by the engagement of chemoattractants with their cognate receptors orchestrate lymphocyte movements into and out of lymphoid organs and sites of inflammation. Yet, the role of chemokines in organizing lymphocyte movements in lymphoid organs is controversial. Recent evidence suggests that the extensive network of fibroblastic reticular cells within the T cell areas helps guide T cells. The expression of adhesion molecules and chemokines by fibroblastic reticular cells most likely facilitates their influence on T cell movements. Consistent with this hypothesis, CD4 T cells with defective chemokine receptor signaling move very differently within lymph nodes than do normal cells. For the imaging studies, we used CD4 T cells prepared from Gnai2(-/-) mice, which lack G(alphai2) expression. We first demonstrate that CD4 as well as CD8 T cells from these mice are markedly defective in chemokine receptor signaling. Gnai2(-/-) T cells have profound defects in chemokine-induced intracellular calcium mobilization, chemotaxis, and homing, whereas Gnai2(+/-) T cells exhibit modest defects. Intravital imaging revealed that within the inguinal lymph nodes Gnai2(-/-) CD4 T accumulate at the cortical ridge, poorly accessing the lymph node paracortex. They also lack the customary amoeboid-like cell movements and active membrane projections observed with normal CD4 T cells. These results demonstrate the importance of G(alphai2) for T lymphocyte chemokine receptor signaling and argue that local chemoattractants regulate the movement of CD4 T cells in lymph nodes.


Assuntos
Inibição de Migração Celular , Quimiotaxia de Leucócito/imunologia , Subunidade alfa Gi2 de Proteína de Ligação ao GTP/biossíntese , Subunidade alfa Gi2 de Proteína de Ligação ao GTP/deficiência , Linfonodos/imunologia , Linfonodos/metabolismo , Subpopulações de Linfócitos T/imunologia , Subpopulações de Linfócitos T/metabolismo , Animais , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/metabolismo , Linfócitos T CD4-Positivos/patologia , Sinalização do Cálcio/genética , Sinalização do Cálcio/imunologia , Quimiotaxia de Leucócito/genética , Regulação para Baixo/genética , Regulação para Baixo/imunologia , Subunidade alfa Gi2 de Proteína de Ligação ao GTP/genética , Subunidade alfa Gi2 de Proteína de Ligação ao GTP/fisiologia , Regulação da Expressão Gênica/imunologia , Linfonodos/patologia , Ativação Linfocitária/genética , Ativação Linfocitária/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Isoformas de Proteínas/biossíntese , Isoformas de Proteínas/deficiência , Isoformas de Proteínas/fisiologia , Receptores de Quimiocinas/antagonistas & inibidores , Receptores de Quimiocinas/biossíntese , Receptores de Quimiocinas/fisiologia , Subpopulações de Linfócitos T/patologia
19.
Immunology ; 122(2): 199-209, 2007 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17490434

RESUMO

Galphai2-deficient mice, which spontaneously develop colitis, have previously been reported to have an increased frequency of mature, single positive thymocytes compared to wild-type mice. In this study we further characterized the intrathymic changes in these mice before and during overt colitis. Even before the onset of colitis, Galphai2(-/-) thymi weighed less and contained fewer thymocytes, and this was exacerbated with colitis development. Whereas precolitic Galphai2(-/-) mice had unchanged thymocyte density compared to Galphai2(+/-) mice of the same age, this was significantly decreased in mice with colitis. Thymic atrophy in Galphai2(-/-) mice involved mainly the cortex. Using a five-stage phenotypic characterization of thymocyte maturation based on expression of CD4, CD8, TCRalphabeta, CD69 and CD62L, we found that both precolitic and colitic Galphai2(-/-) mice had significantly increased frequencies of mature single-positive CD4(+) and CD8(+) medullary thymocytes, and significantly reduced frequencies and total numbers of immature CD4(+) CD8(+) double-positive thymocytes compared to Galphai2(+/-) mice. Furthermore, cortical and transitional precolitic Galphai2(-/-) thymocytes showed significantly reduced chemotactic migration towards CXCL12, and a trend towards reduced migration to CCL25, compared to wild-type thymocytes, a feature even more pronounced in colitic mice. This impaired chemotactic migration of Galphai2(-/-) thymocytes could not be reversed by increased chemokine concentrations. Galphai2(-/-) thymocytes also showed reduced expression of the CCL25 receptor CCR9, but not CXCR4, the receptor, for CXCL12. Finally, wild-type colonic lamina propria lymphocytes migrated in response to CXCL12, but not CCL25 and, as with thymocytes, the chemokine responsiveness was significantly reduced in Galphai2(-/-) mucosal lymphocytes.


Assuntos
Quimiotaxia de Leucócito/imunologia , Colite/imunologia , Subunidade alfa Gi2 de Proteína de Ligação ao GTP/deficiência , Subpopulações de Linfócitos T/imunologia , Timo/imunologia , Animais , Quimiocina CXCL12 , Quimiocinas/imunologia , Quimiocinas CXC/imunologia , Colite/patologia , Colo/imunologia , Modelos Animais de Doenças , Progressão da Doença , Feminino , Subunidade alfa Gi2 de Proteína de Ligação ao GTP/genética , Mucosa Intestinal/imunologia , Contagem de Linfócitos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Tamanho do Órgão , Receptores CCR , Receptores CXCR4/metabolismo , Receptores de Quimiocinas/metabolismo , Timo/patologia
20.
Am J Physiol Gastrointest Liver Physiol ; 292(6): G1511-9, 2007 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-17332478

RESUMO

Mice deficient in the G-protein alpha subunit G(i)alpha(2) spontaneously develop colitis and colon cancer. IL-11 is a pleiotropic cytokine known to protect the intestinal epithelium from injury in animal models of colitis and is produced by subepithelial myofibroblasts in response to inflammatory mediators including TGF-beta, IL-1beta, and PGE(2). Arachidonic acid release and subsequent PGE(2) production is significantly decreased in the colonic mucosa of G(i)alpha(2)-/- mice, and we hypothesized that this would affect mucosal IL-11 production. Mucosal levels of IL-11 were found to be significantly decreased in G(i)alpha(2)-/- mice despite the presence of mild colitis. Primary cultures of G(i)alpha(2)-/- intestinal and colonic myofibroblasts (IMF and CMF, respectively) produced less basal and TGF-beta or IL-1beta-stimulated IL-11 mRNA and protein than wild-type cells. Inhibitors of ERK or p38 MAPK activation dose dependently inhibited IMF and CMF IL-11 production in response to TGF-beta stimulation, whereas 16,16 dimethyl-PGE(2) and prostanoid receptor subtype-selective agonists induced IL-11 production. Treatment of animals with the EP4-specific agonist ONO-AE1-329 resulted in enhanced mucosal levels of IL-11, and increased IL-11 production by ex vivo cultured CMF. Modulation of cAMP levels produced diverging results, with enhancement of TGF-beta-induced IL-11 release in IMF pretreated with 8-Br-cAMP and inhibition in cells treated either with pertussis toxin or the PKA inhibitor H-89. These data suggest a physiological role for prostaglandins, MAPK signaling, and cAMP signaling for the production of myofibroblast-derived IL-11 in the mouse intestinal mucosa.


Assuntos
Colo/metabolismo , Dinoprostona/metabolismo , Fibroblastos/metabolismo , Subunidade alfa Gi2 de Proteína de Ligação ao GTP/metabolismo , Interleucina-11/metabolismo , Intestino Delgado/metabolismo , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Transdução de Sinais , 16,16-Dimetilprostaglandina E2/farmacologia , Alprostadil/análogos & derivados , Alprostadil/farmacologia , Animais , Células Cultivadas , Colo/citologia , Colo/efeitos dos fármacos , Colo/enzimologia , AMP Cíclico/metabolismo , Relação Dose-Resposta a Droga , Ativação Enzimática , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Fibroblastos/efeitos dos fármacos , Fibroblastos/enzimologia , Flavonoides/farmacologia , Subunidade alfa Gi2 de Proteína de Ligação ao GTP/deficiência , Subunidade alfa Gi2 de Proteína de Ligação ao GTP/genética , Imidazóis/farmacologia , Interleucina-11/genética , Interleucina-1beta/metabolismo , Intestino Delgado/citologia , Intestino Delgado/efeitos dos fármacos , Intestino Delgado/enzimologia , Éteres Metílicos/farmacologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas Quinases Ativadas por Mitógeno/antagonistas & inibidores , Inibidores de Proteínas Quinases/farmacologia , Piridinas/farmacologia , RNA Mensageiro/metabolismo , Receptores de Prostaglandina E/metabolismo , Receptores de Prostaglandina E Subtipo EP4 , Transdução de Sinais/efeitos dos fármacos , Fator de Crescimento Transformador beta/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...