Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
2.
Biomed Pharmacother ; 103: 869-878, 2018 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-29710503

RESUMO

Cardiovascular dysfunction is a major complication associated with sepsis induced mortality. Cardiac fibrosis plays a critical role in sepsis induced cardiac dysfunction. The mechanisms of the activation of cardiac fibrosis is unclarified. In this study, we found that microRNA-23b (miR-23b) was up-regulated in heart tissue during cecal ligation and puncture (CLP)-induced sepsis and transfection of miR-23b inhibitor improved survival in late sepsis. Inhibition of miR-23b in the myocardium protected against cardiac output and enhanced left ventricular systolic function. miR-23b inhibitor also alleviated cardiac fibrosis in late sepsis. MiR-23b mediates the activation of TGF-ß1/Smad2/3 signaling to promote the differentiation of cardiac fibroblasts through suppression of 5'TG3'-interacting factor 1 (TGIF1). MiR-23b also induces AKT/N-Cadherin signaling to contribute to the deposition of extracellular matrix by inhibiting phosphatase and tensin homologue (PTEN). TGIF1 and PTEN were confirmed as the targets of miR-23b in vitro by Dual-Glo Luciferase assay. miR-23b inhibitor blocked the activation of adhesive molecules and restored the imbalance of pro-fibrotic and anti-fibrotic factors. These data provide direct evidence that miR-23b is a critical contributor to the activation of cardiac fibrosis to mediate the development of myocardial dysfunction in late sepsis. Blockade of miR-23b expression may be an effective approach for prevention sepsis-induced cardiac dysfunction.


Assuntos
Coração/fisiopatologia , Proteínas de Homeodomínio/metabolismo , MicroRNAs/genética , Miocárdio/patologia , PTEN Fosfo-Hidrolase/metabolismo , Proteínas Repressoras/metabolismo , Sepse/genética , Animais , Fibrose , Células HEK293 , Testes de Função Cardíaca , Humanos , Camundongos Endogâmicos C57BL , MicroRNAs/antagonistas & inibidores , Sepse/patologia , Análise de Sobrevida
3.
J Infect Dis ; 218(2): 300-311, 2018 06 20.
Artigo em Inglês | MEDLINE | ID: mdl-29506272

RESUMO

Background: microRNA-23b (miR-23b) is a multiple functional miRNA. We hypothesize that miR-23b plays a role in the pathogenesis of sepsis. Our study investigated the effect of miR-23b on sepsis-induced immunosuppression. Methods: Mice were treated with miR-23b inhibitors by tail vein injection 2 days after cecal ligation puncture (CLP)-induced sepsis. Apoptosis in spleens and apoptotic signals were investigated, and survival was monitored. T-cell immunoreactivities were examined during late sepsis. Nuclear factor κB (NF-κB)-inducing kinase (NIK), tumor necrosis factor (TNF)-receptor associated factor 1 (TRAF1), and X-linked inhibitor of apoptosis protein (XIAP), the putative targets of miR-23b, were identified by a dual-luciferase reporter assay. Results: miR-23b expression is upregulated and sustained during sepsis. The activation of the TLR4/TLR9/p38 MAPK/STAT3 signal pathway contributes to the production of miR-23b in CLP-induced sepsis. miR-23b inhibitor decreased the number of spleen cells positive by terminal deoxynucleotidyl transferase dUTP nick-end labeling and improved survival. miR-23b inhibitor restored the immunoreactivity by alleviating the development of T-cell exhaustion and producing smaller amounts of immunosuppressive interleukin 10 and interleukin 4 during late sepsis. We demonstrated that miR-23b mediated immunosuppression during late sepsis by inhibiting the noncanonical NF-κB signal and promoting the proapoptotic signal pathway by targeting NIK, TRAF1, and XIAP. Conclusions: Inhibition of miR-23b reduces late-sepsis-induced immunosuppression and improves survival. miR-23b might be a target for immunosuppression.


Assuntos
Tolerância Imunológica , Proteínas Inibidoras de Apoptose/metabolismo , MicroRNAs/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Sepse/patologia , Fator 1 Associado a Receptor de TNF/metabolismo , Animais , Apoptose , Fusão Gênica Artificial , Modelos Animais de Doenças , Perfilação da Expressão Gênica , Genes Reporter , Proteínas Inibidoras de Apoptose/análise , Luciferases/análise , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , MicroRNAs/antagonistas & inibidores , Proteínas Serina-Treonina Quinases/análise , Baço/patologia , Análise de Sobrevida , Linfócitos T/imunologia , Fator 1 Associado a Receptor de TNF/análise , Quinase Induzida por NF-kappaB
4.
J Neuroimmunol ; 314: 30-41, 2018 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-29169800

RESUMO

Depending on the duration and severity, psychological tension and physical stress can enhance or suppress the immune system in both humans and animals. Although it has been established that chronic stress exerts a significant suppressive effect on immune function, the mechanisms by which affects immune responses remain elusive. By employing an in vivo murine system, we revealed that TGF-ß1/Smad2/3/Foxp3 axis was remarkably activated following chronic stress. Furthermore, TLR9 and p38 MAPK played a critical role in the activation of TGF-ß1/Smad2/3/Foxp3 signaling cascade. Moreover, inhibition of TGF-ß1/Smad2/3/Foxp3 or p38 significantly attenuated chronic stress-induced lymphocyte apoptosis and apoptosis-related proteins, as well as the differentiation of T regulatory cells in spleen. Interestingly, disequilibrium of pro-inflammatory and anti-inflammatory cytokines balance caused by chronic stress was also rescued by blocking TGF-ß1/Smad2/3/Foxp3 axis. These findings yield insight into a novel mechanism by which chronic stress modulates immune functions and identifies new targets for the development of novel anti-immune suppressant medications.


Assuntos
Tolerância Imunológica/imunologia , Transdução de Sinais/imunologia , Estresse Psicológico/imunologia , Animais , Fatores de Transcrição Forkhead/imunologia , Fatores de Transcrição Forkhead/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Proteína Smad2/imunologia , Proteína Smad2/metabolismo , Proteína Smad3/imunologia , Proteína Smad3/metabolismo , Estresse Psicológico/metabolismo , Fator de Crescimento Transformador beta1/imunologia , Fator de Crescimento Transformador beta1/metabolismo
5.
J Neuroimmunol ; 309: 72-76, 2017 08 15.
Artigo em Inglês | MEDLINE | ID: mdl-28601292

RESUMO

Physical or psychological chronic stress can suppress the immune system. However, the mechanisms remain to be elucidated. We investigated the effect of hematopoietic stem-progenitor cells (HSPCs) on chronic stress-induced the alterations of immune responses. We demonstrate that HSPCs prevents stress-induced lymphocyte apoptosis. Moreover, we also demonstrate that the protective effect of HSPCs on stress-induced lymphocyte reduction exerts by steroid hormones. Furthermore, we reveal that chronic stress-induced T cell-mediated immune responses contributes to the protective effect of HSPCs. These results indicate that HPSCs might offer a novel therapeutic strategy against the deleterious effects of chronic stress on the immune system.


Assuntos
Apoptose/fisiologia , Transplante de Células-Tronco Hematopoéticas/métodos , Células-Tronco Hematopoéticas/imunologia , Linfócitos/fisiologia , Estresse Psicológico/imunologia , Estresse Psicológico/terapia , Animais , Células Cultivadas , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Estresse Psicológico/psicologia
6.
Oncotarget ; 8(29): 47317-47329, 2017 Jul 18.
Artigo em Inglês | MEDLINE | ID: mdl-28525390

RESUMO

Cardiac dysfunction is correlated with detrimental prognosis of sepsis and contributes to a high risk of mortality. After an initial hyperinflammatory reaction, most patients enter a protracted state of immunosuppression (late sepsis) that alters both innate and adaptive immunity. The changes of cardiac function in late sepsis are not yet known. MicroRNA-155 (miR-155) is previously found to play important roles in both regulations of immune activation and cardiac function. In this study, C57BL/6 mice were operated to develop into early and late sepsis phases, and miR-155 mimic was injected through the tail vein 48 h after cecal ligation and puncture (CLP). The effect of miR-155 on CLP-induced cardiac dysfunction was explored in late sepsis. We found that increased expression of miR-155 in the myocardium protected against cardiac dysfunction in late sepsis evidenced by attenuating sepsis-reduced cardiac output and enhancing left ventricular systolic function. We also observed that miR-155 markedly reduced the infiltration of macrophages and neutrophils into the myocardium and attenuated the inflammatory response via suppression of JNK signaling pathway. Moreover, overexpression of ß-arrestin 2 (Arrb2) exacerbated the mice mortality and immunosuppression in late sepsis. Furthermore, transfection of miR-155 mimic reduced Arrb2 expression, and then restored immunocompetence and improved survival in late septic mice. We conclude that increased miR-155 expression through systemic administration of miR-155 mimic attenuates cardiac dysfunction and improves late sepsis survival by targeting JNK associated inflammatory signaling and Arrb2 mediated immunosuppression.


Assuntos
Cardiopatias/etiologia , Cardiopatias/metabolismo , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , MicroRNAs/genética , Sepse/complicações , beta-Arrestina 2/metabolismo , Animais , Citocinas/metabolismo , Modelos Animais de Doenças , Expressão Gênica , Regulação da Expressão Gênica , Cardiopatias/mortalidade , Cardiopatias/fisiopatologia , Testes de Função Cardíaca , Humanos , Mediadores da Inflamação/metabolismo , Macrófagos/metabolismo , Masculino , Camundongos , Miocárdio/metabolismo , Neutrófilos/metabolismo , Interferência de RNA , Sepse/etiologia , Transdução de Sinais , Taxa de Sobrevida , Transfecção
7.
Biochem Biophys Rep ; 7: 130-137, 2016 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-27957549

RESUMO

Sepsis is an exaggerated systemic inflammatory response to persistent bacteria infection with high morbidity and mortality rate clinically. ß-arrestin 2 modulates cell survival and cell death in different systems. However, the effect of ß-arrestin 2 on sepsis-induced cardiac dysfunction is not yet known. Here, we show that ß-arrestin 2 overexpression significantly enhances animal survival following cecal ligation and puncture (CLP)-induced sepsis. Importantly, overexpression of ß-arrestin 2 in mice prevents CLP-induced cardiac dysfunction. Also, ß-arrestin 2 overexpression dramatically attenuates CLP-induced myocardial gp130 and p38 mitogen-activated protein kinase (MAPK) phosphorylation levels following CLP. Therefore, ß-arrestin 2 prevents CLP-induced cardiac dysfunction through gp130 and p38. These results suggest that modulation of ß-arrestin 2 might provide a novel therapeutic approach to prevent cardiac dysfunction in patients with sepsis.

8.
Apoptosis ; 21(5): 546-57, 2016 May.
Artigo em Inglês | MEDLINE | ID: mdl-26921179

RESUMO

Elevated circulatory free fatty acids (FFAs) especially saturated FFAs, such as palmitate (PA), are detrimental to the heart. However, mechanisms responsible for this phenomenon remain unknown. Here, the role of JAK2/STAT3 in PA-induced cytotoxicity was investigated in cardiomyocytes. We demonstrate that PA suppressed the JAK2/STAT3 pathway by dephosphorylation of JAK2 (Y1007/1008) and STAT3 (Y705), and thus blocked the translocation of STAT3 into the nucleus. Conversely, phosphorylation of S727, another phosphorylated site of STAT3, was increased in response to PA treatment. Pretreatment of JNK inhibitor, but not p38 MAPK inhibitor, inhibited STAT3 (S727) activation induced by PA and rescued the phosphorylation of STAT3 (Y705). The data suggested that JNK may be another upstream factor regulating STAT3, and verified the important function of P-STAT3 (Y705) in PA-induced cardiomyocyte apoptosis. Sodium orthovanadate (SOV), a protein tyrosine phosphatase inhibitor, obviously inhibited PA-induced apoptosis by restoring JAK2/STAT3 pathways. This effect was diminished by STAT3 inhibitor Stattic. Collectively, our data suggested a novel mechanism that the inhibition of JAK2/STAT3 activation was responsible for palmitic lipotoxicity and SOV may act as a potential therapeutic agent by targeting JAK2/STAT3 in lipotoxic cardiomyopathy treatment.


Assuntos
Apoptose/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Janus Quinase 2/metabolismo , Miócitos Cardíacos/efeitos dos fármacos , Palmitatos/toxicidade , Fator de Transcrição STAT3/metabolismo , Vanadatos/farmacologia , Animais , Linhagem Celular , Células Cultivadas , Janus Quinase 2/antagonistas & inibidores , Miócitos Cardíacos/enzimologia , Miócitos Cardíacos/metabolismo , Palmitatos/antagonistas & inibidores , Fosforilação/efeitos dos fármacos , Proteínas Tirosina Fosfatases/antagonistas & inibidores , Ratos Sprague-Dawley , Fator de Transcrição STAT3/antagonistas & inibidores , Transdução de Sinais/efeitos dos fármacos
9.
J Biol Chem ; 291(2): 605-12, 2016 Jan 08.
Artigo em Inglês | MEDLINE | ID: mdl-26582201

RESUMO

Recent studies reveal that multifunctional protein ß-arrestin 2 (Arrb2) modulates cell apoptosis. Survival and various aspects of liver injury were investigated in WT and Arrb2 KO mice after bile duct ligation (BDL). We found that deficiency of Arrb2 enhances survival and attenuates hepatic injury and fibrosis. Following BDL, Arrb2-deficient mice as compared with WT controls displayed a significant reduction of hepatocyte apoptosis as demonstrated by the TUNEL assay. Following BDL, the levels of phospho-Akt and phospho-glycogen synthase kinase 3ß (GSK3ß) in the livers were significantly increased in Arrb2 KO compared with WT mice, although p-p38 increased in WT but not in Arrb2-deficient mice. Inhibition of GSK3ß following BDL decreases hepatic apoptosis and decreased p-p38 in WT mice but not in Arrb2 KO mice. Activation of Fas receptor with Jo2 reduces phospho-Akt and increases apoptosis in WT cells and WT mice but not in Arrb2-deficient cells and Arrb2-deficient mice. Consistent with direct interaction of Arrb2 with and regulating Akt phosphorylation, the expression of a full-length or N terminus but not the C terminus of Arrb2 reduces Akt phosphorylation and coimmunoprecipates with Akt. These results reveal that the protective effect of deficiency of Arrb2 is due to loss of negative regulation of Akt due to BDL and decreased downstream GSK3ß and p38 MAPK signaling pathways.


Assuntos
Apoptose , Arrestinas/metabolismo , Hepatócitos/patologia , Proteínas Proto-Oncogênicas c-akt/antagonistas & inibidores , Animais , Arrestinas/deficiência , Ductos Biliares/metabolismo , Ativação Enzimática , Quinase 3 da Glicogênio Sintase/antagonistas & inibidores , Quinase 3 da Glicogênio Sintase/metabolismo , Glicogênio Sintase Quinase 3 beta , Ligadura , Fígado/enzimologia , Fígado/patologia , Cirrose Hepática/metabolismo , Cirrose Hepática/patologia , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fosforilação , Proteínas Proto-Oncogênicas c-akt/metabolismo , Análise de Sobrevida , beta-Arrestina 2 , beta-Arrestinas , Receptor fas/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
10.
PLoS One ; 10(4): e0123447, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25885582

RESUMO

Emerging evidence implied that chronic stress has been exerting detrimental impact on immune system functions in both humans and animals. Toll-like receptors (TLRs) have been shown to play an essential role in modulating immune responses and cell survival. We have recently shown that TLR9 deficiency protects against lymphocyte apoptosis induced by chronic stress. However, the exact role of TLR9 in stress-mediated change of macrophage function remains unclear. The results of the current study showed that when BALB/c mice were treated with restraint stress (12 h daily for 2 days), the number of macrophages recruited to the peritoneal cavity was obviously increased. Results also demonstrated that the sustained effects of stress elevated cytokine IL-1ß, TNF-α and IL-10 production yet diminished IFN-γ production from macrophage, which led to apoptotic cell death. However, TLR9 deficiency prevented the chronic stress-mediated accumulation of macrophages. In addition, knocking out TLR9 significantly abolished the chronic stress-induced imbalance of cytokine levels and apoptosis in macrophage. TLR9 deficiency was also found to reverse elevation of plasma IL-1ß, IL-10 and IL-17 levels and decrease of plasma IFN-γ level under the condition of chronic stress. These results indicated that TLR9-mediated macrophage responses were required for chronic stress-induced immunosuppression. Further exploration showed that TLR9 deficiency prevented the increment of p38 MAPK phosphorylation and reduction of Akt/Gsk-3ß phosphorylation; TLR9 deficiency also attenuated the release of mitochondrial cytochrome c into cytoplasm, caused upregulation of Bcl-2/Bax protein ratio, downregulation of cleavage of caspase-3 and PARP, as well as decreased TUNEL-positive cells in macrophage of stressed mice. Collectively, our studies demonstrated that deficiency of TLR9 maintained macrophage function by modulating macrophage accumulation and attenuating macrophage apoptosis, thus preventing immunosuppression in restraint-stressed mice.


Assuntos
Apoptose/fisiologia , Citocinas/metabolismo , Macrófagos/metabolismo , Estresse Fisiológico/fisiologia , Receptor Toll-Like 9/metabolismo , Animais , Caspase 3/metabolismo , Regulação para Baixo , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Restrição Física , Receptor Toll-Like 9/genética
11.
Cell Immunol ; 295(2): 92-8, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25880099

RESUMO

Sepsis, a major clinical problem with high morbidity and mortality, is caused by overwhelming systemic host-inflammatory response. Toll-like receptors (TLRs) play a fundamental role in induction of hyperinflammation and tissue damage in sepsis. In this study, we demonstrate a protective role of TLR9 inhibition against the dysregulated inflammatory response and tissue injury in sepsis. TLR9 deficiency decreased the mortality of mice following cecal ligation and puncture (CLP)-induced sepsis. TLR9 knockout mice showed dampened p38 activation and augmented Akt phosphorylation in the spleen, lung and liver. In addition, TLR9 deficiency decreased the levels of inflammatory cytokines and attenuated splenic apoptosis after CLP. These results indicate that TLR9 inhibition might offer a novel therapeutic strategy for the management of sepsis.


Assuntos
Sepse/imunologia , Receptor Toll-Like 9/antagonistas & inibidores , Animais , Apoptose/imunologia , Citocinas/genética , Citocinas/imunologia , Marcação In Situ das Extremidades Cortadas , Estimativa de Kaplan-Meier , Fígado/imunologia , Pulmão/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Proteína Oncogênica v-akt/imunologia , RNA/química , RNA/genética , Reação em Cadeia da Polimerase em Tempo Real , Sepse/patologia , Baço/imunologia , Receptor Toll-Like 9/genética , Receptor Toll-Like 9/imunologia , Proteínas Quinases p38 Ativadas por Mitógeno/imunologia
12.
J Immunol ; 194(3): 1021-30, 2015 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-25539811

RESUMO

Opioids have been widely applied in clinics as one of the most potent pain relievers for centuries, but their abuse has deleterious physiological effects including immunosuppression. However, the mechanisms are unclear. TLRs and acetylcholine are widely expressed in the immune and nervous systems, and play critical roles in immune responses. In this article, we show that morphine suppresses the innate immunity in microglia and bone marrow-derived macrophages through differential regulation of TLRs and acetylcholinesterase. Either morphine or inhibition of acetylcholine significantly promotes upregulation of microRNA-124 (miR-124) in microglia, bone marrow-derived macrophages, and the mouse brain, where miR-124 mediates morphine inhibition of the innate immunity by directly targeting a subunit of NF-κB p65 and TNFR-associated factor 6 (TRAF6). Furthermore, transcription factors AP-1 and CREB inhibited miR-124, whereas p65 bound directly to promoters of miR-124, thereby enhancing miR-124 transcription. Moreover, acute morphine treatment transiently upregulated the expression of p65 and phospho-p65 in both nucleus and cytoplasm priming the expression of miR-124, whereas long exposure of morphine maintained miR-124 expression, which inhibited p65- and TRAF6-dependent TLR signaling. These data suggest that modulation of miRs is capable of preventing opioid-induced damage to microglia.


Assuntos
Imunomodulação/genética , MicroRNAs/genética , Microglia/imunologia , Microglia/metabolismo , Interferência de RNA , Fator 6 Associado a Receptor de TNF/genética , Fator de Transcrição RelA/genética , Regiões 3' não Traduzidas , Animais , Pareamento de Bases , Sequência de Bases , Sítios de Ligação , Linhagem Celular , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Imunidade Inata/efeitos dos fármacos , Imunidade Inata/genética , Masculino , Camundongos , Microglia/efeitos dos fármacos , Morfina/farmacologia , Transcrição Gênica
13.
J Biol Chem ; 289(33): 23075-23085, 2014 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-25012660

RESUMO

The control of IL-10 production in Toll-like receptor (TLR) signals remains to be elucidated. Here, we report that ß-arrestin 2 positively regulates TLR-triggered IL-10 production in a p38 mitogen-activated protein kinase (MAPK)-dependent mechanism. In vitro studies with cells including peritoneal macrophages and HEK293/TLR4 cells have demonstrated that ß-arrestin 2 forms complexes with p38 and facilitates p38 activation after lipopolysaccharide (LPS) stimulation. Deficiency of ß-arrestin 2 and inhibition of p38 MAPK activity both ameliorate TLR4-stimulated IL-10 response. Additionally, in vivo experiments show that mice lacking ß-arrestin 2 produce less amount of IL-10, and are more susceptible to LPS-induced septic shock which is further enhanced by blocking IL-10 signal. These results reveal a novel mechanism by which ß-arrestin 2 negatively regulates TLR4-mediated inflammatory reactions.


Assuntos
Arrestinas/metabolismo , Interleucina-10/metabolismo , Sistema de Sinalização das MAP Quinases , Macrófagos Peritoneais/metabolismo , Receptor 4 Toll-Like/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Animais , Arrestinas/genética , Células HEK293 , Humanos , Inflamação/genética , Inflamação/metabolismo , Inflamação/patologia , Interleucina-10/genética , Lipopolissacarídeos/toxicidade , Macrófagos Peritoneais/patologia , Camundongos , Camundongos Knockout , Choque Séptico/induzido quimicamente , Choque Séptico/genética , Choque Séptico/metabolismo , Choque Séptico/patologia , Receptor 4 Toll-Like/genética , beta-Arrestina 2 , beta-Arrestinas , Proteínas Quinases p38 Ativadas por Mitógeno/genética
14.
Brain Behav Immun ; 36: 118-27, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24513872

RESUMO

Stress can either enhance or suppress immune functions depending on a variety of factors such as duration of stressful condition. Chronic stress has been demonstrated to exert a significant suppressive effect on immune function. However, the mechanisms responsible for this phenomenon remain to be elucidated. Here, male C57BL/6 mice were placed in a 50-ml conical centrifuge tube with multiple punctures to establish a chronic restraint stress model. Serum IL-10 levels, IL-10 production by the splenocytes, and activation of STAT3 in the mouse spleen were assessed. We demonstrate that IL-10/STAT3 axis was remarkably activated following chronic stress. Moreover, TLR4 and p38 MAPK play a pivotal role in the activation of IL-10/STAT3 signaling cascade. Interestingly, blocking antibody against IL-10 receptor and inhibition of STAT3 by STAT3 inhibitor S3I-201 attenuates stress-induced lymphocyte apoptosis. Inhibition of IL-10/STAT3 dramatically inhibits stress-induced reduction in IL-12 production. Furthermore, disequilibrium of Th1/Th2 cytokine balance caused by chronic stress was also rescued by blocking IL-10/STAT3 axis. These results yield insight into a new mechanism by which chronic stress regulates immune functions. IL-10/STAT3 pathway provides a novel relevant target for the manipulation of chronic stress-induced immune suppression.


Assuntos
Interleucina-10/metabolismo , Fator de Transcrição STAT3/metabolismo , Estresse Psicológico/imunologia , Animais , Interleucina-10/sangue , Sistema de Sinalização das MAP Quinases , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fosforilação , Restrição Física , Baço/metabolismo , Baço/patologia , Estresse Psicológico/metabolismo , Receptor 4 Toll-Like/genética , Receptor 4 Toll-Like/metabolismo
15.
Neuroimmunomodulation ; 21(1): 1-7, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24080854

RESUMO

OBJECTIVES: Mental and physical stress can suppress the immune system in both humans and animals. The mechanism by which stress affects immune responses, however, remains poorly defined. Toll-like receptors (TLRs) play a key role in modulating immune responses and cell survival. The mechanisms by which TLRs modulate chronic stress are largely unexplored. METHODS: Six- to 8-week-old male mice were subjected to chronic 12-hour daily physical restraint stress. Apoptotic cells were determined by the terminal deoxynucleotidyl transferase-mediated deoxyuridine triphosphate nick end labeling (TUNEL) assay. We examined cytokine levels by enzyme-linked immunosorbent Assay (ELISA). The expression of CYP11A1 was determined by quantitative real-time RT-PCR. RESULTS: TLR9-deficient mice were resistant to chronic stress-induced lymphocyte apoptosis. In addition, in TLR9 knockout (KO) mice, chronic stress-induced upregulation of corticosterone levels was significantly decreased. Notably, lymphocytes from both TLR9 KO and wild-type mice were similarly sensitive to corticosteroid-induced cell apoptosis. Moreover, TLR9 deficiency blocked the chronic stress-induced imbalance in T helper (Th) 1 and Th2 cytokine levels. CONCLUSION: Taken together, our findings reveal that TLR9 plays an essential role in chronic stress-induced immune suppression.


Assuntos
Tolerância Imunológica/genética , Baço/metabolismo , Estresse Psicológico/imunologia , Estresse Psicológico/patologia , Receptor Toll-Like 9/fisiologia , Animais , Apoptose/genética , Linfócitos T CD4-Positivos/metabolismo , Enzima de Clivagem da Cadeia Lateral do Colesterol/genética , Enzima de Clivagem da Cadeia Lateral do Colesterol/metabolismo , Corticosterona/sangue , Citocinas/metabolismo , Ensaio de Imunoadsorção Enzimática , Regulação da Expressão Gênica/genética , Tolerância Imunológica/fisiologia , Marcação In Situ das Extremidades Cortadas , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Restrição Física/efeitos adversos , Baço/patologia , Estresse Psicológico/sangue , Estresse Psicológico/etiologia , Estresse Psicológico/genética , Receptor Toll-Like 9/deficiência
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...