Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 176
Filtrar
1.
Circ Heart Fail ; 17(3): e010569, 2024 03.
Artigo em Inglês | MEDLINE | ID: mdl-38410978

RESUMO

BACKGROUND: Exercise training can promote cardiac rehabilitation, thereby reducing cardiovascular disease mortality and hospitalization rates. MicroRNAs (miRs) are closely related to heart disease, among which miR-574-3p plays an important role in myocardial remodeling, but its role in exercise-mediated cardioprotection is still unclear. METHODS: A mouse myocardial hypertrophy model was established by transverse aortic coarctation, and a 4-week swimming exercise training was performed 1 week after the operation. After swimming training, echocardiography was used to evaluate cardiac function in mice, and histopathologic staining was used to detect cardiac hypertrophy, myocardial fibrosis, and cardiac inflammation. Quantitative real-time polymerase chain reaction was used to detect the expression levels of miR-574-3p and cardiac hypertrophy markers. Western blotting detected the IL-6 (interleukin-6)/JAK/STAT inflammatory signaling pathway. RESULTS: Echocardiography and histochemical staining found that aerobic exercise significantly improved pressure overload-induced myocardial hypertrophy (n=6), myocardial interstitial fibrosis (n=6), and cardiac inflammation (n=6). Quantitative real-time polymerase chain reaction detection showed that aerobic exercise upregulated the expression level of miR-574-3p (n=6). After specific knockdown of miR-574-3p in mouse hearts with adeno-associated virus 9 using cardiac troponin T promoter, we found that the protective effect of exercise training on the heart was significantly reversed. Echocardiography and histopathologic staining showed that inhibiting the expression of miR-574-3p could partially block the effects of aerobic exercise on cardiac function (n=6), cardiomyocyte cross-sectional area (n=6), and myocardial fibrosis (n=6). Western blotting and immunohistochemical staining showed that the inhibitory effects of aerobic exercise on the IL-6/JAK/STAT pathway and cardiac inflammation were partially abolished after miR-574-3p knockdown. Furthermore, we also found that miR-574-3p exerts cardioprotective effects in cardiomyocytes by targeting IL-6 (n=3). CONCLUSIONS: Aerobic exercise protects cardiac hypertrophy and inflammation induced by pressure overload by upregulating miR-574-3p and inhibiting the IL-6/JAK/STAT pathway.


Assuntos
Insuficiência Cardíaca , MicroRNAs , Miocardite , Camundongos , Animais , Interleucina-6/metabolismo , Janus Quinases/metabolismo , Insuficiência Cardíaca/metabolismo , Transdução de Sinais , Fatores de Transcrição STAT/metabolismo , Miócitos Cardíacos/metabolismo , MicroRNAs/genética , MicroRNAs/metabolismo , Cardiomegalia/patologia , Miocardite/genética , Miocardite/prevenção & controle , Inflamação/patologia , Modelos Animais de Doenças , Fibrose
2.
J Am Heart Assoc ; 12(4): e028442, 2023 02 21.
Artigo em Inglês | MEDLINE | ID: mdl-36752267

RESUMO

Background Viral myocarditis is characterized by leukocyte infiltration of the heart and cardiomyocyte death. We recently identified C-C chemokine ligand (CCL) 17 as a proinflammatory effector of C-C chemokine receptor 2-positive macrophages and dendritic cells that are recruited to the heart and contribute to adverse left ventricular remodeling following myocardial infarction and pressure overload. Methods and Results Mouse encephalomyocarditis virus was used to investigate the function of CCL17 in a viral myocarditis model. Ccl17Gfp reporter and knockout mice were used to identify the cell types that express CCL17 and delineate the functional importance of CCL17 in encephalomyocarditis virus clearance and myocardial inflammation. Cardiac CCL17 was expressed in C-C chemokine receptor 2-positive macrophages and dendritic cells following encephalomyocarditis virus infection. Colony-stimulating factor 2 (granulocyte-macrophage colony-stimulating factor) signaling was identified as a key regulator of CCL17 expression. Ccl17 deletion resulted in impaired encephalomyocarditis virus clearance, increased cardiomyocyte death, and higher mortality during infection early stage, and aggravated hypertrophy and fibrotic responses in infection long-term stage. An increased abundance of regulatory T cells was detected in the myocardium of injured Ccl17-deficient mice. Depletion of regulatory T cells in Ccl17-deficient mice abrogated the detrimental role of CCL17 deletion by restoring interferon signaling. Conclusions Collectively, these findings identify CCL17 as an important mediator of the host immune response during cardiac viral infection early stage and suggest that CCL17 targeted therapies should be avoided in acute viral myocarditis.


Assuntos
Miocardite , Viroses , Camundongos , Animais , Miocardite/genética , Miocardite/prevenção & controle , Linfócitos T Reguladores , Macrófagos/metabolismo , Camundongos Knockout , Receptores de Quimiocinas/metabolismo , Quimiocina CCL17/metabolismo
3.
BMC Cardiovasc Disord ; 22(1): 69, 2022 02 23.
Artigo em Inglês | MEDLINE | ID: mdl-35196979

RESUMO

BACKGROUND: Myocarditis is a cardiomyopathy associated with the inflammatory response. Rosuvastatin (RS) demonstrates cardioprotective effect in the clinical setting, although its cellular and molecular mechanisms in ameliorating myocarditis are largely unknown. MG53 (muscle-specific E3 ligase Mitsugumin 53), a newly identified striated muscle-specific protein, is involved in skeletal muscle membrane repair. We aimed to explore whether RS mediated the repair of cardiomyocytes in an MG53-dependent manner. METHODS: The RS-induced upregulation of MG53 was determined using RT-qPCR and western blotting. A lipopolysaccharide (LPS)-induced cell inflammatory model was constructed using rat cardiac muscle cell H9C2. Inflammatory injury was evaluated according to the alterations of cell viability, mitochondrial membrane potential, cell apoptosis, and expression of pro-inflammatory cytokines (interleukin-1ß, interleukin-6, tumor necrosis factor-α, and monocyte chemoattractant protein-1). Small interfering RNAs (siRNAs) were used to silence MG53. The cardioprotective effect of RS and the inhibition of this protection by MG53 silence were evaluated in the forementioned in vitro model. The underlying mechanism was finally investigated using western blotting to detected the expressions of apoptotic markers (Bcl-2, Bax, Cleaved caspase-9, Cleaved caspase-3), cell cycle regulatory factors (Cyclin A, Cyclin E1, Cyclin D1, CDK2), and components involved in NF-κB signaling pathway (p-IκBa, Iκba, p-p65, p65). RESULTS: RS ameliorated LPS-induced inflammatory injury. RS upregulated the expression of MG53. MG53 was crucial for the RS-mediated repair response in vitro. Ablation of MG53 inhibited the RS-mediated protective effect. Furthermore, RS and MG53 interact in multiple signaling pathways to modulate recovery. CONCLUSION: RS exerts cardioprotective effect in an MG53-dependent manner. MG53 may serve as a novel drug target for myocarditis treatment.


Assuntos
Lipopolissacarídeos , Miocardite , Animais , Humanos , Lipopolissacarídeos/toxicidade , Proteínas Musculares/metabolismo , Miocardite/patologia , Miocardite/prevenção & controle , Miócitos Cardíacos/metabolismo , NF-kappa B/metabolismo , Ratos , Rosuvastatina Cálcica/farmacologia
4.
Sci Rep ; 11(1): 12432, 2021 06 14.
Artigo em Inglês | MEDLINE | ID: mdl-34127684

RESUMO

Coxsackievirus B3 (CVB3), is commonly implicated in myocarditis, which can lead to dilated cardiomyopathy, in addition to causing acute pancreatitis and meningitis. Yet, no vaccines are currently available to prevent this infection. Here, we describe the derivation of a live attenuated vaccine virus, termed mutant (Mt) 10, encoding a single amino acid substitution H790A within the viral protein 1, that prevents CVB3 infection in mice and protects from both myocarditis and pancreatitis in challenge studies. We noted that animals vaccinated with Mt 10 developed virus-neutralizing antibodies, predominantly containing IgG2a and IgG2b, and to a lesser extent IgG3 and IgG1. Furthermore, by using major histocompatibility complex class II dextramers and tetramers, we demonstrated that Mt 10 induces antigen-specific T cell responses that preferentially produce interferon-γ. Finally, neither vaccine recipients nor those challenged with the wild-type virus revealed evidence of autoimmunity or cardiac injury as determined by T cell response to cardiac myosin and measurement of circulating cardiac troponin I levels, respectively. Together, our data suggest that Mt 10 is a vaccine candidate that prevents CVB3 infection through the induction of neutralizing antibodies and antigen-specific T cell responses, the two critical components needed for complete protection against virus infections in vaccine studies.


Assuntos
Infecções por Coxsackievirus/prevenção & controle , Enterovirus Humano B/imunologia , Miocardite/prevenção & controle , Pancreatite/prevenção & controle , Vacinas Virais/imunologia , Animais , Anticorpos Neutralizantes/sangue , Anticorpos Neutralizantes/imunologia , Antígenos Virais/imunologia , Sítios de Ligação/genética , Proteína de Membrana Semelhante a Receptor de Coxsackie e Adenovirus/metabolismo , Infecções por Coxsackievirus/virologia , Modelos Animais de Doenças , Enterovirus Humano B/genética , Feminino , Humanos , Imunogenicidade da Vacina/genética , Masculino , Camundongos , Mutação , Miocardite/virologia , Pancreatite/virologia , Linfócitos T/imunologia , Vacinas Atenuadas/administração & dosagem , Vacinas Atenuadas/genética , Vacinas Atenuadas/imunologia , Vacinas Virais/administração & dosagem , Vacinas Virais/genética
5.
Zhongguo Fei Ai Za Zhi ; 24(6): 394-403, 2021 Jun 20.
Artigo em Chinês | MEDLINE | ID: mdl-34157799

RESUMO

BACKGROUND: Immune checkpoint inhibitors (ICIs) such as antibodies against programmed cell death 1 (PD-1) and programmed cell death ligand 1 (PD-L1), have shown remarkable efficacies in many subtypes of cancers. However, ICIs may also cause severe immune-related adverse events in the recipient patients. Recently, ICI-associated myocarditis have been reported in hundreds of patients worldwide, with a mortality rate of approximately 50% in these cases. This study aims to recapitulate the cardiotoxicity and explore the detoxicifying approaches to attenuate mortality caused by PD-1/PD-L1 inhibitors in healthy mice. METHODS: Six to eight-week-old C57BL/6 mice were inoculated with anti-PD-1 antibody (12.5 µg/g every 5 days for 6 injections), anti-PD-L1 antibody (10 µg/g once a week for 6 weeks), anti-PD-L1 antibody (with the same dosage described above) in combination with levothyroxine (0.25 µg/g, intraperitoneally injected half an hour before anti-PD-L1 antibody injection), or isotype control immunoglobulin IgG (10 µg/g once a week for 6 weeks). The ejection function of the hearts was detected by echocardiography, body temperature and blood pressure were detected by Mouse MonitorTM and non-invassive blood pressure minotor, and serum free thyroxine concentration was detected by The enzyme linked immunosorbent assay (ELISA). RESULTS: PD-L1 was expressed at different levels by the cardiomyocytes of the mice. The isotype control immunoglobulin and anti-PD-1 antibody did not cause death of the mice. The 12 mice receiving 3-6 injections of anti-PD-L1 antibody showed a significant increase in the heart-to-tibial ratio and cardiomyoctye degeneration, hyalinization and extravascular inflammatory cell infiltration. In addition, the serum thyroxine was mardedly decreased to 1/3 of that in the control group mice, and the blood pressure and body temperature were abnormally decreased in mice upon treatment with PD-L1 blockade. Eight of the 12 (66.7%) mice died from multiple intravenous injection of anti-PD-L1 antibody.Intraperitoneal injection of levothyroxine 30 min before the injection of anti-PD-L1 antibody significantly attenuated the mortality rate of the anti-PD-L1 antibody-treated mice. CONCLUSIONS: The anti-PD-L1 antibody is cardiotoxic and lethal, and levothyroxine is able to rescue the mice from this immune checkpoint inhibitor-caused mortality.


Assuntos
Cardiotoxicidade , Inibidores de Checkpoint Imunológico/efeitos adversos , Tiroxina , Animais , Antígeno B7-H1/antagonistas & inibidores , Cardiotoxicidade/etiologia , Cardiotoxicidade/mortalidade , Cardiotoxicidade/prevenção & controle , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Miocardite/induzido quimicamente , Miocardite/mortalidade , Miocardite/prevenção & controle , Receptor de Morte Celular Programada 1/metabolismo , Hormônios Tireóideos/farmacologia , Hormônios Tireóideos/uso terapêutico , Tiroxina/farmacologia , Tiroxina/uso terapêutico
6.
Front Immunol ; 11: 1955, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33013849

RESUMO

Background: The eosinophilic inflammation plays a critical role in myocarditis (Mcd); its underlying mechanism remains to be further elucidated. This study aims to investigate the role of Bcl2-like protein 12 (Bcl2L12) in inducing the defects of apoptosis in eosinophils (Eos) of the heart tissues. Methods: Human explant heart samples were collected. Eosinophilia and myocarditis (Mcd)-like inflammation were induced in the mouse heart by immunizing with murine cardiac α-myosin heavy chain (MyHCα) peptides. Results: Markedly more Eos were observed in heart tissues from patients with Mcd than those from patients with dilated cardiomyopathy. Eos isolated from Mcd hearts showed the signs of apoptosis defects. The Eo counts in the Mcd heart tissues were positively correlated with the Bcl2L12 expression in Eos isolated from the heart tissues. Exposure to interleukin 5 in the culture induced the expression of Bcl2L12 in Eos. Bcl2L12 bound c-Myc, the transcription factor of Fas ligand (FasL), to prevent c-Myc from binding to the FasL promoter, to restrict the FasL gene transcription in Eos. Inhibition of Bcl2L12 prevented the induction of eosinophilia and Mcd-like inflammation in the mouse heart. Conclusions: The Bcl2L12 expression contributes to apoptosis defects in Eos of the Mcd heart. Blocking Bcl2L12 prevents the eosinophilia induction and alleviates Mcd-like inflammation in mice.


Assuntos
Apoptose/efeitos dos fármacos , Eosinofilia/prevenção & controle , Eosinófilos/efeitos dos fármacos , Proteínas Musculares/metabolismo , Miocardite/prevenção & controle , Proteínas Proto-Oncogênicas c-bcl-2/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Adulto , Animais , Células Cultivadas , Modelos Animais de Doenças , Eosinofilia/genética , Eosinofilia/imunologia , Eosinofilia/metabolismo , Eosinófilos/imunologia , Eosinófilos/metabolismo , Proteína Ligante Fas/metabolismo , Feminino , Humanos , Interleucina-5/metabolismo , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas Musculares/genética , Miocardite/genética , Miocardite/imunologia , Miocardite/metabolismo , Cadeias Pesadas de Miosina , Fragmentos de Peptídeos , Proteínas Proto-Oncogênicas c-bcl-2/genética , Interferência de RNA , Transdução de Sinais , Adulto Jovem
7.
J Cell Mol Med ; 24(18): 10677-10692, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32757377

RESUMO

Heart failure (HF) represents a major public health burden. Inflammation has been shown to be a critical factor in the progression of HF, regardless of the aetiology. Disappointingly, the majority of clinical trials targeting aspects of inflammation in patients with HF have been largely negative. Many clinical researches demonstrate that danshen has a good efficacy on HF, and however, whether danshen exerts anti-inflammatory effects against HF remains unclear. In our study, the employment of a water extracted and alcohol precipitated of danshen extract attenuated cardiac dysfunction and inflammation response in acute myocardial infarction-induced HF rats. Transcriptome technique and validation results revealed that TLR4 signalling pathway was involved in the anti-inflammation effects of danshen. In vitro, danshen reduced the release of inflammatory mediators in LPS-stimulated RAW264.7 macrophage cells. Besides, the LPS-stimulated macrophage conditioned media was applied to induce cardiac H9C2 cells injury, which could be attenuated by danshen. Furtherly, knock-down and overexpression of TLR4 were utilized to confirm that danshen ameliorated inflammatory injury via MyD88-dependent TLR4-TRAF6-NF-κB signalling pathway in cardiomyocytes. Furthermore, by utilizing co-immunoprecipitation, danshen was proved to suppress MD2/TLR4 complex formation and MyD88 recruitment. In conclusion, our results demonstrated that danshen ameliorates inflammatory injury by controlling MD2/TLR4-MyD88 complex formation and TLR4-TRAF6-NF-κB signalling pathway in acute myocardial infarction-induced HF.


Assuntos
Insuficiência Cardíaca/tratamento farmacológico , Antígeno 96 de Linfócito/antagonistas & inibidores , Fator 88 de Diferenciação Mieloide/antagonistas & inibidores , Infarto do Miocárdio/complicações , Fitoterapia , Extratos Vegetais/uso terapêutico , Salvia miltiorrhiza/química , Transdução de Sinais/efeitos dos fármacos , Receptor 4 Toll-Like/antagonistas & inibidores , Animais , Biomarcadores , Meios de Cultivo Condicionados/farmacologia , Avaliação Pré-Clínica de Medicamentos , Insuficiência Cardíaca/genética , Insuficiência Cardíaca/prevenção & controle , Antígeno 96 de Linfócito/fisiologia , Macrófagos/metabolismo , Camundongos , Complexos Multiproteicos/efeitos dos fármacos , Fator 88 de Diferenciação Mieloide/fisiologia , Miocardite/prevenção & controle , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/metabolismo , Extratos Vegetais/isolamento & purificação , Células RAW 264.7 , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/farmacologia , Ratos , Ratos Sprague-Dawley , Proteínas Recombinantes/metabolismo , Transdução de Sinais/genética , Organismos Livres de Patógenos Específicos , Receptor 4 Toll-Like/fisiologia , Transcriptoma/efeitos dos fármacos , Disfunção Ventricular Esquerda/etiologia , Disfunção Ventricular Esquerda/prevenção & controle
8.
ESC Heart Fail ; 7(5): 2838-2852, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32662949

RESUMO

AIMS: Left ventricular (LV) dysfunction in viral myocarditis is attributed to myocardial inflammation and fibrosis, inducing acute and long-time cardiac damage. Interventions are not established. On the basis of the link between inflammation, fibrosis, aldosterone, and extracellular matrix regulation, we aimed to investigate the effect of an early intervention with the mineralocorticoid receptor antagonist (MRA) eplerenone on cardiac remodelling in a murine model of persistent coxsackievirus B3 (CVB3)-induced myocarditis. METHODS AND RESULTS: SWR/J mice were infected with 5 × 104 plaque-forming units of CVB3 (Nancy strain) and daily treated either with eplerenone (200 mg/kg body weight) or with placebo starting from Day 1. At Day 8 or 28 post infection, mice were haemodynamically characterized and subsequently sacrificed for immunohistological and molecular biology analyses. Eplerenone did not influence CVB3 load. Already at Day 8, 1.8-fold (P < 0.05), 1.4-fold (P < 0.05), 3.2-fold (P < 0.01), and 2.1-fold (P < 0.001) reduction in LV intercellular adhesion molecule 1 expression, presence of monocytes/macrophages, oxidative stress, and apoptosis, respectively, was observed in eplerenone-treated vs. untreated CVB3-infected mice. In vitro, eplerenone led to 1.4-fold (P < 0.01) and 1.2-fold (P < 0.01) less CVB3-induced cardiomyocyte oxidative stress and apoptosis. Furthermore, collagen production was 1.1-fold (P < 0.05) decreased in cardiac fibroblasts cultured with medium of eplerenone-treated vs. untreated CVB3-infected HL-1 cardiomyocytes. These ameliorations were in vivo translated into prevention of cardiac fibrosis, as shown by 1.4-fold (P < 0.01) and 2.1-fold (P < 0.001) lower collagen content in the LV of eplerenone-treated vs. untreated CVB3-infected mice at Days 8 and 28, respectively. This resulted in an early and long-lasting improvement of LV dimension and function, as indicated by reduced LV end-systolic volume and end-diastolic volume, and an increase in LV contractility (dP/dtmax ) and LV relaxation (dP/dtmin ), respectively (P < 0.05). CONCLUSIONS: Early intervention with the MRA eplerenone modulates the acute host and defence reaction and prevents cardiac disease progression in experimental CVB3-induced myocarditis without aggravation of viral load. The findings advocate for an initiation of therapy of viral myocarditis as early as possible, even before the onset of inflammation-induced myocardial dysfunction. This may also have implications for coronavirus disease-19 therapy.


Assuntos
Fibrose Endomiocárdica/prevenção & controle , Enterovirus Humano B/patogenicidade , Eplerenona/farmacologia , Miocardite/tratamento farmacológico , Miocardite/virologia , Disfunção Ventricular Esquerda/virologia , Análise de Variância , Animais , Biópsia por Agulha , Modelos Animais de Doenças , Progressão da Doença , Fibrose Endomiocárdica/patologia , Imuno-Histoquímica , Masculino , Metaloproteinases da Matriz/efeitos dos fármacos , Metaloproteinases da Matriz/metabolismo , Camundongos , Camundongos Transgênicos , Miocardite/prevenção & controle , Distribuição Aleatória , Valores de Referência , Resultado do Tratamento , Disfunção Ventricular Esquerda/patologia , Disfunção Ventricular Esquerda/fisiopatologia
9.
Leukemia ; 34(7): 1726-1729, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32483300

RESUMO

The scientific community faces an unexpected and urgent challenge related to the SARS-CoV-2 pandemic and is investigating the role of receptors involved in entry of this virus into cells as well as pathomechanisms leading to a cytokine "storm," which in many cases ends in severe acute respiratory syndrome, fulminant myocarditis and kidney injury. An important question is if it may also damage hematopoietic stem progenitor cells?


Assuntos
Infecções por Coronavirus/epidemiologia , Síndrome da Liberação de Citocina/epidemiologia , Células-Tronco Hematopoéticas/virologia , Inflamassomos/imunologia , Pandemias , Pneumonia Viral/epidemiologia , Síndrome Respiratória Aguda Grave/epidemiologia , Injúria Renal Aguda/epidemiologia , Injúria Renal Aguda/imunologia , Injúria Renal Aguda/prevenção & controle , Injúria Renal Aguda/virologia , Enzima de Conversão de Angiotensina 2 , Betacoronavirus/efeitos dos fármacos , Betacoronavirus/imunologia , Betacoronavirus/patogenicidade , COVID-19 , Infecções por Coronavirus/tratamento farmacológico , Infecções por Coronavirus/imunologia , Infecções por Coronavirus/virologia , Síndrome da Liberação de Citocina/imunologia , Síndrome da Liberação de Citocina/prevenção & controle , Síndrome da Liberação de Citocina/virologia , Citocinas/antagonistas & inibidores , Citocinas/genética , Citocinas/imunologia , Furanos/farmacologia , Regulação da Expressão Gênica , Células-Tronco Hematopoéticas/efeitos dos fármacos , Células-Tronco Hematopoéticas/imunologia , Compostos Heterocíclicos de 4 ou mais Anéis , Humanos , Imunidade Inata/efeitos dos fármacos , Fatores Imunológicos/farmacologia , Indenos , Inflamassomos/antagonistas & inibidores , Inflamassomos/genética , Miocardite/epidemiologia , Miocardite/imunologia , Miocardite/prevenção & controle , Miocardite/virologia , Proteína 3 que Contém Domínio de Pirina da Família NLR/antagonistas & inibidores , Proteína 3 que Contém Domínio de Pirina da Família NLR/genética , Proteína 3 que Contém Domínio de Pirina da Família NLR/imunologia , Peptidil Dipeptidase A/genética , Peptidil Dipeptidase A/imunologia , Pneumonia Viral/tratamento farmacológico , Pneumonia Viral/imunologia , Pneumonia Viral/virologia , Piroptose/efeitos dos fármacos , Piroptose/genética , Piroptose/imunologia , Fatores de Risco , SARS-CoV-2 , Síndrome Respiratória Aguda Grave/imunologia , Síndrome Respiratória Aguda Grave/prevenção & controle , Síndrome Respiratória Aguda Grave/virologia , Glicoproteína da Espícula de Coronavírus/genética
10.
Circ Heart Fail ; 13(6): e006573, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-32493060

RESUMO

BACKGROUND: Previous studies have shown beneficial effects of acute infusion of the primary ketone body, ß-hydroxybutyrate, in heart failure (HF). However, whether chronic elevations in circulating ketones are beneficial remains unknown. METHODS: To chronically elevate circulating ketones in mice, we deleted the expression of the ketolytic, rate-limiting-enzyme, SCOT (succinyl-CoA:3-ketoacid-CoA transferase 1; encoded by Oxct1), in skeletal muscle. Tamoxifen-inducible skeletal muscle-specific Oxct1Muscle-/- knockout (n=32) mice and littermate controls (wild type; WT; n=35) were subjected to transverse aortic constriction (TAC) surgery to induce HF. RESULTS: Deletion of SCOT in skeletal, but not cardiac muscle resulted in elevated concentrations of fasted circulating ß-hydroxybutyrate in knockout mice compared with WT mice (P=0.030). Five weeks following TAC, WT mice progressed to HF, whereas knockout mice with elevated fasting circulating ketones were largely protected from the TAC-induced effects observed in WT mice (ejection fraction, P=0.011; mitral E/A, P=0.012). Furthermore, knockout mice with TAC had attenuated expression of markers of sterile inflammation and macrophage infiltration, which were otherwise elevated in WT mice subjected to TAC. Lastly, addition of ß-hydroxybutyrate to isolated hearts was associated with reduced NLRP3 (nucleotide-binding domain-like receptor protein 3)-inflammasome activation, which has been previously shown to play a role in contributing to HF-induced cardiac inflammation. CONCLUSIONS: These data show that chronic elevation of circulating ketones protects against the development of HF that is associated with the ability of ß-hydroxybutyrate to directly reduce inflammation. These beneficial effects of ketones were associated with reduced cardiac NLRP3 inflammasome activation, suggesting that ketones may modulate cardiac inflammation via this mechanism.


Assuntos
Ácido 3-Hidroxibutírico/sangue , Coenzima A-Transferases/deficiência , Insuficiência Cardíaca/prevenção & controle , Miocardite/prevenção & controle , Miocárdio/enzimologia , Animais , Coenzima A-Transferases/genética , Modelos Animais de Doenças , Fibrose , Insuficiência Cardíaca/sangue , Insuficiência Cardíaca/enzimologia , Insuficiência Cardíaca/fisiopatologia , Hipertrofia Ventricular Esquerda/sangue , Hipertrofia Ventricular Esquerda/fisiopatologia , Hipertrofia Ventricular Esquerda/prevenção & controle , Inflamassomos/metabolismo , Preparação de Coração Isolado , Masculino , Camundongos Knockout , Miocardite/sangue , Miocardite/enzimologia , Miocardite/fisiopatologia , Miocárdio/patologia , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Regulação para Cima , Disfunção Ventricular Esquerda/sangue , Disfunção Ventricular Esquerda/fisiopatologia , Disfunção Ventricular Esquerda/prevenção & controle , Função Ventricular Esquerda , Remodelação Ventricular
11.
Biosci Rep ; 40(6)2020 06 26.
Artigo em Inglês | MEDLINE | ID: mdl-32515469

RESUMO

BACKGROUND: Genistein is widely used as a pharmacological compound as well as a food additive. However, the pharmaceutical effects of Genistein on myocarditis and its potential mechanisms have not been studied in detail. METHODS: H9c2 cells were continuously stimulated by lipopolysaccharide (LPS) for 12 h to simulate the in vitro model of myocarditis injury. DrugBank, String, and GEO dataset were used to investigate specific genes that interacting with Genistein. KEGG and GO enrichment analysis were employed to explore Myc-related signaling pathways. Biological behaviors of H9c2 cells were observed with the support of cell counting kit-8, MTT and flow cytometry. Expression levels of cytokines including TNF-α and ILs were evaluated by enzyme-linked immunosorbent assay. Western blot was applied to detect the expression of Myc and MAPK pathway related proteins. RESULTS: Genistein alleviated the damage of H9c2 cells subjected to LPS from the perspective of elevating cells growth ability, and inhibiting cells apoptosis and inflammatory response. Through bioinformatics analysis, we identified Myc as the potential target of Genistein in myocarditis, and MAPK as the signaling pathway. Significantly, Myc was highly up-regulated in myocarditis samples. More importantly, by performing biological experiments, we discovered that Genistein relieved H9c2 cells apoptosis and inflammatory reaction which caused by LPS stimulation through inhibiting Myc expression. Additionally, the marked augmentation of p-P38 MAPK and p-JNK expression in LPS-induced cardiomyocyte model were blocked by Genistein and si-Myc. CONCLUSIONS: Our research revealed that Myc mediated the protective effects of Genistein on H9c2 cells damage caused by LPS partly through modulation of MAPK/JNK signaling pathway.


Assuntos
Anti-Inflamatórios/farmacologia , Genisteína/farmacologia , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Lipopolissacarídeos/toxicidade , Miocardite/prevenção & controle , Miócitos Cardíacos/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-myc/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Animais , Apoptose/efeitos dos fármacos , Linhagem Celular , Proliferação de Células/efeitos dos fármacos , Citocinas/metabolismo , Mediadores da Inflamação/metabolismo , Miocardite/enzimologia , Miocardite/patologia , Miócitos Cardíacos/enzimologia , Miócitos Cardíacos/patologia , Fosforilação , Ratos , Transdução de Sinais
12.
Int J Biol Sci ; 16(11): 1798-1810, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32398950

RESUMO

Aims: The High Mobility Group A1 (HMGA1) proteins, serving as a dynamic regulator of gene transcription and chromatin remodeling, play an influential part in the pathological process of a large number of cardiovascular diseases. However, the precise role of HMGA1 in sepsis induced cardiomyopathy (SIC) remains unintelligible. This research was designed to illustrate the effect of HMGA1 involved in SIC. Methods and Results: Cardiomyocyte-specific HMGA1 overexpression was obtained using an adeno-associated virus system with intramyocardial injection in mice heart. The model of SIC in mice was constructed via intraperitoneal injection of lipopolysaccharide (LPS) for 6h. H9c2 rat cardiomyocytes was stimulated with LPS for 12h. HMGA1 expression was upregulated in murine inflammatory hearts as well as LPS stimulated H9c2 cardiomyocytes. HMGA1-overexpressing exhibited aggravated cardiac dysfunction, cardiac inflammation as well as cells apoptosis following LPS treatment both in vivo and in vitro experiment. Interestingly, HMGA1 knockdown in H9c2 cardiomyocytes attenuated LPS-induced cardiomyocyte inflammation, but aggravated cell apoptosis. Mechanistically, we found that overexpression of HMGA1 induced increased expression of cyclooxygenase-2 (COX-2). COX-2 inhibitor alleviated the aggravation of inflammation and apoptosis in HMGA1 overexpressed H9c2 cardiomyocytes whereas HMGA1 knockdown induced a reduction in signal transducer and activators of transcription 3 (STAT3) expression. STAT3 agonist reversed HMGA1 silence induced anti-inflammatory effects, while ameliorated cell apoptosis induced by LPS. Conclusion: In conclusion, our results suggest that overexpression of HMGA1 aggravated cardiomyocytes inflammation and apoptosis by up-regulating COX-2 expression, while silence of HMGA1 expression attenuated inflammation but aggregated cell apoptosis via down-regulation of STAT3.


Assuntos
Proteína HMGA1a/metabolismo , Lipopolissacarídeos/toxicidade , Miocardite/induzido quimicamente , Miocardite/prevenção & controle , Miócitos Cardíacos/efeitos dos fármacos , Animais , Cardiomiopatias/metabolismo , Cardiomiopatias/patologia , Linhagem Celular , Citocinas/genética , Citocinas/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Proteína HMGA1a/genética , Inflamação/induzido quimicamente , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Miocardite/metabolismo , Miocárdio/metabolismo , Miocárdio/patologia , Miócitos Cardíacos/metabolismo , Ratos
13.
Cell Immunol ; 347: 104025, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31837749

RESUMO

B10 cells, a specific subset of regulatory B cells, are capable of regulating immune response and restricting inflammation and autoimmune disease progression by producing IL-10. B10 cells frequently change significantly during inflammation and autoimmunity. However, how B10 cell populations change in viral myocarditis (VMC) remains unclear. Therefore, this work was conducted to clarify the changes in B10 cells and their potential mechanisms. Our results showed that the B10 cell frequency significantly changed in the VMC model. Changes in prostaglandin E2 (PGE2) levels in VMC model hearts were consistent with B10 expansion. PGE2 induced B10 cell expansion via the MAPKs/AKT-AP1 axis or AhR signaling. Additionally, PGE2-pretreated B10 cells inhibited naïve CD4+ T cell differentiation into Th17 cells. In vivo, PGE2 treatment or adoptive B10 cell transfer significantly restricted VMC development. Our results provide sufficient evidence that PGE2-induced B10 cell expansion may become a promising therapeutic approach for VMC and acute inflammatory injury.


Assuntos
Linfócitos B Reguladores/imunologia , Dinoprostona/farmacologia , Infecções por Enterovirus/patologia , Interleucina-10/imunologia , Miocardite/prevenção & controle , Células Th17/citologia , Transferência Adotiva , Animais , Subpopulações de Linfócitos B/imunologia , Diferenciação Celular/efeitos dos fármacos , Células Cultivadas , Dinoprostona/sangue , Enterovirus Humano B , Ativação Linfocitária/imunologia , Contagem de Linfócitos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Miocardite/tratamento farmacológico , Miocardite/virologia , Miocárdio/imunologia , Peptídeos , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais/imunologia , Células Th17/imunologia
14.
JCI Insight ; 4(16)2019 08 22.
Artigo em Inglês | MEDLINE | ID: mdl-31434807

RESUMO

Mesenchymal stem cells (MSCs) can suppress pathological inflammation. However, the mechanisms underlying the association between MSCs and inflammation remain unclear. Under coculture conditions with macrophages, MSCs highly expressed angiopoietin-like 4 (ANGPTL4) to blunt the polarization of macrophages toward the proinflammatory phenotype. ANGPTL4-deficient MSCs failed to inhibit the inflammatory macrophage phenotype. In inflammation-related animal models, the injection of coculture medium or ANGPTL4 protein increased the antiinflammatory macrophages in both peritonitis and myocardial infarction. In particular, cardiac function and pathology were markedly improved by ANGPTL4 treatment. We found that retinoic acid-related orphan receptor α (RORα) was increased by inflammatory mediators, such as IL-1ß, and bound to ANGPTL4 promoter in MSCs. Collectively, RORα-mediated ANGPTL4 induction was shown to contribute to the antiinflammatory activity of MSCs against macrophages under pathological conditions. This study suggests that the capability of ANGPTL4 to induce tissue repair is a promising opportunity for safe stem cell-free regeneration therapy from a translational perspective.


Assuntos
Proteína 4 Semelhante a Angiopoietina/fisiologia , Ativação de Macrófagos , Macrófagos/metabolismo , Células-Tronco Mesenquimais/metabolismo , Infarto do Miocárdio/terapia , Peritonite/terapia , Proteína 4 Semelhante a Angiopoietina/genética , Proteína 4 Semelhante a Angiopoietina/metabolismo , Animais , Anti-Inflamatórios não Esteroides , Polaridade Celular , Células Cultivadas , Técnicas de Cocultura , Modelos Animais de Doenças , Humanos , Inflamação/imunologia , Inflamação/terapia , Mediadores da Inflamação/metabolismo , Macrófagos/imunologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Infarto do Miocárdio/imunologia , Miocardite/etiologia , Miocardite/prevenção & controle , Membro 1 do Grupo F da Subfamília 1 de Receptores Nucleares/metabolismo , Peritonite/imunologia , Receptores do Ácido Retinoico/metabolismo
15.
J Mol Histol ; 50(5): 405-415, 2019 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-31256303

RESUMO

Viral myocarditis has been found to be one of the leading causes of sudden death in young adults. However, no effective drugs have been developed to intervene the progression of myocarditis. Accordingly, the present study is carried out to explore the protective role played by melatonin in the setting of viral myocarditis with a focus on Mst1-Hippo pathway, mitochondrial dysfunction and ER stress. Cardiac function was determined via echocardiographic examination. Mitochondrial function and ER stress were detected via ELISA, western blots, and immunofluorescence. Our data demonstrated that virus injection induced cardiac dysfunction as evidenced by reduced contractile function in myocardium. Besides, LDH release assay and western blotting analysis demonstrated that cardiomyocyte death was activated by virus injection. Interestingly, melatonin treatment improved cardiac function and repressed virus-mediated cardiomyocyte apoptosis. At the molecular levels, mitochondrial dysfunction was induced by virus infection, as indicated by mitochondrial membrane potential reduction, mPTP opening rate elevation and caspase-9-related apoptosis activation. Besides, ER stress parameters were also elevated in virus-treated cardiomyocytes. Interestingly, melatonin treatment maintained mitochondrial dysfunction and repressed ER stress. To the end, we found that Mst1 was upregulated by virus infection; this effect was attenuated through supplementation with melatonin. However, Mst1 overexpression reduced the beneficial impact exerted by melatonin on cardiomyocyte viability, mitochondrial function and ER homeostasis. Our study illustrated that melatonin treatment attenuated viral myocarditis via sustaining cardiomyocyte viability, repressing mitochondrial dysfunction and inhibiting ER stress in a manner dependent on Mst1 inhibition.


Assuntos
Estresse do Retículo Endoplasmático/efeitos dos fármacos , Fator de Crescimento de Hepatócito/antagonistas & inibidores , Melatonina/farmacologia , Mitocôndrias/efeitos dos fármacos , Miocardite/prevenção & controle , Proteínas Proto-Oncogênicas/antagonistas & inibidores , Animais , Sobrevivência Celular/efeitos dos fármacos , Fator de Crescimento de Hepatócito/metabolismo , Humanos , Mitocôndrias/patologia , Miócitos Cardíacos/efeitos dos fármacos , Substâncias Protetoras/farmacologia , Proteínas Proto-Oncogênicas/metabolismo , Vírus/patogenicidade
16.
J Nutr Biochem ; 71: 63-71, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-31284167

RESUMO

Dietary omega-3 polyunsaturated fatty acids (n-3 PUFAs) have been associated with a lower incidence of cardiovascular events and sudden cardiac death. Flaxseed is a rich plant source of n-3 PUFAs and can retard the progression and accelerate the regression of atherosclerotic plaques. The aim of the study was to examine the preventive and therapeutic effects of dietary flaxseed on arrhythmias and heart dysfunction that develops after a myocardial infarction (MI). The left anterior descending coronary artery was ligated in rats to induce the MI. Rats were randomized into five groups: sham MI with normal chow, MI with normal chow, MI with 10% milled flaxseed supplementation (flax), MI with 4.4% supplemented flax oil enriched in alpha-linolenic acid (ALA) and MI with flax lignan secoisolariciresinol diglucoside (SDG) supplementation (0.44%). Animals were fed with their respective diets for 2 weeks before and for 8 weeks after the surgery. Echocardiography and continuous electrocardiographic recordings were obtained after ligation to confirm the induction of the MI, to check for arrhythmias and to assess cardiac function. Histological examination was also performed to evaluate cardiac fibrosis. Dietary supplementation with flaxseed, ALA or SDG before and after the induction of the MI significantly reduced the incidence of arrhythmias and resulted in significantly smaller infarct size, less left ventricle dilation, and decreased myocardial fibrosis and tumor necrosis factor-α levels compared to the control MI group. Together, this study supports a beneficial effect of dietary flaxseed in patients for the prevention and treatment of arrhythmias and ventricular remodeling post-MI.


Assuntos
Arritmias Cardíacas/prevenção & controle , Cardiotônicos/farmacologia , Linho/química , Infarto do Miocárdio/complicações , Animais , Arritmias Cardíacas/diagnóstico por imagem , Arritmias Cardíacas/etiologia , Peso Corporal/efeitos dos fármacos , Butileno Glicóis/farmacologia , Cardiotônicos/química , Suplementos Nutricionais , Eletrocardiografia , Ácidos Graxos/sangue , Glucosídeos/farmacologia , Masculino , Infarto do Miocárdio/fisiopatologia , Miocardite/etiologia , Miocardite/patologia , Miocardite/prevenção & controle , Tamanho do Órgão/efeitos dos fármacos , Ratos Sprague-Dawley , Remodelação Ventricular , Ácido alfa-Linolênico/farmacologia
17.
Am J Physiol Heart Circ Physiol ; 317(3): H531-H540, 2019 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-31225989

RESUMO

As an inflammatory disease afflicting the heart muscle, autoimmune myocarditis (AM) represents one of the foremost causes of heart failure. Accumulating evidence has implicated microRNAs (miRNAs) in the process of inflammation and autoimmunity. Hence, the current study aimed to investigate the mechanism by which miR-141-3p influences experimental AM (EAM). An EAM mouse model was established using 6-wk old male BALB/c mice, after which the expression of miR-141-3p and STAT4 was measured. Gain-of-function and loss-of-function investigations were performed to identify the functional role of miR-141-3p and STAT4 in EAM. Heart weight-to-body weight ratio, cardiac function, and degree of inflammation, as well as the levels of inflammation factors (IFN-γ, TNF-α, IL-2, IL-6, and IL-17) in the serum were detected. STAT4 was subsequently verified to be upregulated, and miR-141-3p was downregulated in the EAM mice. Furthermore, the overexpression of miR-141-3p or silencing of STAT4 was observed to reduce the heart weight-to-body weight ratio of EAM mice and improve cardiac function, while alleviating the degree of inflammatory cell infiltration in the myocardial tissue. Meanwhile, the overexpression of miR-141-3p was identified to diminish serum inflammatory factor levels by downregulating STAT4. Additionally, miR-141-3p could bind to STAT4 to downregulate its expression, ultimately mitigating inflammation and inducing an anti-inflammatory effect in EAM mice. Taken together, upregulation of miR-141-3p alleviates the inflammatory response in EAM mice by inhibiting STAT4, providing a promising intervention target for the molecular treatment of AM.NEW & NOTEWORTHY miR-141-3p is poorly expressed, and STAT4 is upregulated in experimental autoimmune myocarditis (EAM) mice. Overexpressing miR-141-3p inhibits EAM. miR-141-3p binds to and suppresses STAT4 expression. miR-141-3p overexpression inhibits inflammatory factors by downregulating STAT4. This study provides new insights into the treatment of autoimmune myocarditis.


Assuntos
Doenças Autoimunes/prevenção & controle , MicroRNAs/metabolismo , Miocardite/prevenção & controle , Miocárdio/metabolismo , RNA Interferente Pequeno/administração & dosagem , Terapêutica com RNAi , Fator de Transcrição STAT4/metabolismo , Animais , Antagomirs/administração & dosagem , Doenças Autoimunes/genética , Doenças Autoimunes/imunologia , Doenças Autoimunes/metabolismo , Citocinas/sangue , Citocinas/genética , Modelos Animais de Doenças , Redes Reguladoras de Genes , Mediadores da Inflamação/sangue , Masculino , Camundongos Endogâmicos BALB C , MicroRNAs/genética , Miocardite/genética , Miocardite/imunologia , Miocardite/metabolismo , Miocárdio/imunologia , Miosinas , Interferência de RNA , Fator de Transcrição STAT4/genética
18.
Inflammation ; 42(4): 1179-1189, 2019 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-30848408

RESUMO

Signal transducer and activator of transcription 4 (STAT4) has been implicated in the progression of myocarditis. The aim of the current study was to investigate the role by which STAT4 influences autoimmune myocarditis in an attempt to identify a theoretical therapeutic perspective for the condition. After successful establishment of an autoimmune myocarditis rat model, the expression patterns of STAT4, NF-κB pathway-related genes, Th1 inflammatory cytokines (IFN-γ and IL-2), and Th2 inflammatory cytokines (IL-6 and IL-10) were subsequently determined. The rats with autoimmune myocarditis were treated with oe-STAT4 or sh-STAT4 lentiviral vectors to evaluate the role of STAT4 in autoimmune myocarditis, or administrated with 1 mL 10 µmol/L of BAY11-7082 (the NF-κB pathway inhibitor) via tail vein to investigate the effect of the NF-κB pathway on autoimmune myocarditis. Finally, cell apoptosis was evaluated. The serum levels of IFN-γ and IL-2, extent of IκBα and P65 phosphorylation, and the expression of STAT4 were elevated, while the serum levels of IL-6 and IL-10 as well as the expression of IκBα were reduced among the rats with autoimmune myocarditis, which was accompanied by an increase in the apoptotic cells. More importantly, the silencing of STAT4 or the inhibition of the NF-κB pathway was detected to result in a decrease in the serum levels of IFN-γ and IL-2 and an elevation of the serum levels of IL-6 and IL-10, and inhibited myocardial cell apoptosis in rats with autoimmune myocarditis. Moreover, STAT4 silencing was also observed to decrease the extent of IκBα and P65 phosphorylation while acting to elevate the expression of IκBα. Taken together, silencing of STAT4 could hinder the progression of autoimmune myocarditis by balancing the expression of Th1/Th2 inflammatory cytokines via the NF-κB pathway, which may provide a novel target for experimental autoimmune myocarditis (EAM) treatment.


Assuntos
Miocardite/imunologia , NF-kappa B/antagonistas & inibidores , Fator de Transcrição STAT4/fisiologia , Células Th1/imunologia , Células Th2/imunologia , Animais , Apoptose , Doenças Autoimunes , Citocinas/sangue , Progressão da Doença , Miocardite/prevenção & controle , NF-kappa B/metabolismo , Ratos , Fator de Transcrição STAT4/antagonistas & inibidores , Fator de Transcrição STAT4/metabolismo
19.
J Cardiovasc Pharmacol ; 73(3): 186-194, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30839512

RESUMO

OBJECTIVE: To investigate whether phenylephrine (PE) inhibits sepsis-induced cardiac dysfunction, cardiac inflammation, and mitochondrial injury through the PI3K/Akt signaling pathway. METHODS: A rat model of sepsis was established by cecal ligation and puncture. PE and/or wortmannin (a PI3K inhibitor) were administered to investigate the role of PI3K/Akt signaling in mediating the effects of PE on inhibiting sepsis-induced cardiac dysfunction, cardiac inflammation, and mitochondrial injury. Hematoxylin-eosin staining, echocardiography, and Langendorff system were used to examine the myocardial injury and function. The concentrations of TNF-α and IL-6 were analyzed by enzyme-linked immunosorbent assay. Intercellular cell adhesion molecule-1 (ICAM-1), vascular cell adhesion molecule-1 (VCAM-1), myeloperoxidase, mitochondria-related fusion/fission proteins, and PI3K/Akt signaling pathway-associated proteins were analyzed by Western blotting. RESULTS: PE improved the cardiac function and survival in septic rats. PE decreased TNF-α, IL-6, ICAM-1, VCAM-1, and myeloperoxidase contents in the myocardium of septic rats. Meanwhile, PE increased the fusion-related proteins and decreased the fission-related proteins in the myocardial mitochondria of septic rats. On the other hand, PE activated the PI3K/Akt signaling pathway in the cecal ligation and puncture-treated rats, and all the protective effects of PE were abolished by wortmannin. CONCLUSIONS: PE attenuated sepsis-induced cardiac dysfunction, cardiac inflammation, and mitochondrial injury through the PI3K/Akt signaling pathway.


Assuntos
Mitocôndrias Cardíacas/efeitos dos fármacos , Dinâmica Mitocondrial/efeitos dos fármacos , Miocardite/prevenção & controle , Miócitos Cardíacos/efeitos dos fármacos , Fenilefrina/farmacologia , Fosfatidilinositol 3-Quinase/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Sepse/tratamento farmacológico , Animais , Modelos Animais de Doenças , Mediadores da Inflamação/metabolismo , Preparação de Coração Isolado , Masculino , Mitocôndrias Cardíacas/enzimologia , Mitocôndrias Cardíacas/patologia , Proteínas Mitocondriais/metabolismo , Miocardite/enzimologia , Miocardite/etiologia , Miocardite/patologia , Miócitos Cardíacos/enzimologia , Miócitos Cardíacos/patologia , Peroxidase/metabolismo , Ratos Sprague-Dawley , Sepse/complicações , Transdução de Sinais , Volume Sistólico/efeitos dos fármacos , Função Ventricular Esquerda
20.
J Cell Physiol ; 234(10): 18029-18040, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-30843214

RESUMO

Fingolimod (FTY720) after phosphorylation, as the ligand of sphingosine 1-phosphate receptors (S1PRs), plays an important role in cell proliferation and differentiation. In this article, FTY720 in the treatment of coxsackievirus B3 (CVB3)-induced viral myocarditis was closely related to apoptosis and AKT/caspase-3 apoptotic pathways. We found that CVB3 inhibited myocardial apoptosis at the early stage with upregulating p-AKT level and downregulating activated caspase-3 level for replication of virus progeny, whereas it promoted apoptosis at a late stage with downregulating p-AKT and upregulating activated caspase-3 for releasing the newly synthesized virus to spread. Interestingly, FTY720 could reverse this trend; it promoted apoptosis at an early stage and inhibited apoptosis at the late stage in vivo and vitro, which proved the antiviral effect. We also found that S1PR1, S1PR4, and S1PR5, rather than S1PR2 and S1PR3, were regulated by FTY720 in this process. The results confirmed that FTY720 alleviates CVB3-induced myocarditis and inhibits viral replication through regulating S1PRs and AKT/caspase-3 pathways with a bidirectional regulation of apoptosis.


Assuntos
Antivirais/farmacologia , Caspase 3/metabolismo , Infecções por Coxsackievirus/prevenção & controle , Enterovirus Humano B/efeitos dos fármacos , Cloridrato de Fingolimode/farmacologia , Miocardite/prevenção & controle , Miócitos Cardíacos/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-akt/metabolismo , Moduladores do Receptor de Esfingosina 1 Fosfato/farmacologia , Receptores de Esfingosina-1-Fosfato/efeitos dos fármacos , Replicação Viral/efeitos dos fármacos , Animais , Apoptose/efeitos dos fármacos , Células Cultivadas , Infecções por Coxsackievirus/metabolismo , Infecções por Coxsackievirus/patologia , Infecções por Coxsackievirus/virologia , Modelos Animais de Doenças , Enterovirus Humano B/crescimento & desenvolvimento , Interações Hospedeiro-Patógeno , Masculino , Camundongos Endogâmicos BALB C , Miocardite/metabolismo , Miocardite/patologia , Miocardite/virologia , Miócitos Cardíacos/enzimologia , Miócitos Cardíacos/patologia , Miócitos Cardíacos/virologia , Transdução de Sinais , Receptores de Esfingosina-1-Fosfato/metabolismo , Fatores de Tempo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA