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.
Acta Pharmacol Sin ; 45(4): 728-737, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38086898

RESUMO

Stimulation of adult cardiomyocyte proliferation is a promising strategy for treating myocardial infarction (MI). Earlier studies have shown increased CCL2 levels in plasma and cardiac tissue both in MI patients and mouse models. In present study we investigated the role of CCL2 in cardiac regeneration and the underlying mechanisms. MI was induced in adult mice by permanent ligation of the left anterior descending artery, we showed that the serum and cardiac CCL2 levels were significantly increased in MI mice. Intramyocardial injection of recombinant CCL2 (rCCL2, 1 µg) immediately after the surgery significantly promoted cardiomyocyte proliferation, improved survival rate and cardiac function, and diminished scar sizes in post-MI mice. Alongside these beneficial effects, we observed an increased angiogenesis and decreased cardiomyocyte apoptosis in post-MI mice. Conversely, treatment with a selective CCL2 synthesis inhibitor Bindarit (30 µM) suppressed both CCL2 expression and cardiomyocyte proliferation in P1 neonatal rat ventricle myocytes (NRVMs). We demonstrated in NRVMs that the CCL2 stimulated cardiomyocyte proliferation through STAT3 signaling: treatment with rCCL2 (100 ng/mL) significantly increased the phosphorylation levels of STAT3, whereas a STAT3 phosphorylation inhibitor Stattic (30 µM) suppressed rCCL2-induced cardiomyocyte proliferation. In conclusion, this study suggests that CCL2 promotes cardiac regeneration via activation of STAT3 signaling, underscoring its potential as a therapeutic agent for managing MI and associated heart failure.


Assuntos
Insuficiência Cardíaca , Infarto do Miocárdio , Humanos , Camundongos , Animais , Ratos , Quimiocina CCL2/metabolismo , Infarto do Miocárdio/metabolismo , Miócitos Cardíacos , Insuficiência Cardíaca/metabolismo , Regeneração , Camundongos Endogâmicos C57BL , Apoptose , Fator de Transcrição STAT3/metabolismo
2.
J Mol Cell Cardiol ; 187: 26-37, 2024 02.
Artigo em Inglês | MEDLINE | ID: mdl-38150867

RESUMO

Stimulating cardiomyocyte proliferation in the adult heart has emerged as a promising strategy for cardiac regeneration following myocardial infarction (MI). The NRG1-ERBB4 signaling pathway has been implicated in the regulation of cardiomyocyte proliferation. However, the therapeutic potential of recombinant human NRG1 (rhNRG1) has been limited due to the low expression of ERBB4 in adult cardiomyocytes. Here, we investigated whether a fusion protein of rhNRG1 and an ERBB3 inhibitor (rhNRG1-HER3i) could enhance the affinity of NRG1 for ERBB4 and promote adult cardiomyocyte proliferation. In vitro and in vivo experiments were conducted using postnatal day 1 (P1), P7, and adult cardiomyocytes. Western blot analysis was performed to assess the expression and activity of ERBB4. Cardiomyocyte proliferation was evaluated using Ki67 and pH 3 immunostaining, while fibrosis was assessed using Masson staining. Our results indicate that rhNRG1-HER3i, but not rhNRG1, promoted P7 and adult cardiomyocyte proliferation. Furthermore, rhNRG1-HER3i improved cardiac function and reduced cardiac fibrosis in post-MI hearts. Administration of rhNRG1-HER3i inhibited ERBB3 phosphorylation while increasing ERBB4 phosphorylation in adult mouse hearts. Additionally, rhNRG1-HER3i enhanced angiogenesis following MI compared to rhNRG1. In conclusion, our findings suggest that rhNRG1-HER3i is a viable therapeutic approach for promoting adult cardiomyocyte proliferation and treating MI by enhancing NRG1-ERBB4 signaling pathway.


Assuntos
Cardiomiopatias , Infarto do Miocárdio , Camundongos , Animais , Humanos , Transdução de Sinais , Miócitos Cardíacos/metabolismo , Neuregulina-1/uso terapêutico , Cardiomiopatias/metabolismo , Receptor ErbB-4/metabolismo
3.
Int Heart J ; 63(5): 814-820, 2022 Sep 30.
Artigo em Inglês | MEDLINE | ID: mdl-36104232

RESUMO

Nonalcoholic fatty liver disease (NAFLD) is an emerging driver of cardiac arrhythmias. However, the relationship between NAFLD and malignant arrhythmia in non-ST-segment elevation myocardial infarction (NSTEMI) patients is still unclear.In this study, 358 NSTEMI inpatients were enrolled. They all received 24-hour Holter monitoring after percutaneous coronary intervention. All inpatients were divided into two groups: the non-NAFLD group (236 cases, 65.9%) and the NAFLD group (122 cases, 34.1%). Compared with the non-NAFLD group, the NAFLD group had a significantly higher incidence of PVCs/hour > 5 (premature ventricular complexes, 32.0% versus 9.3%, P < 0.001), ventricular tachycardia (VT, 22.1% versus 5.9%, P < 0.001), and sinus arrest (SA, 7.4% versus 1.3%, P = 0.002). We found that NAFLD was closely associated with the occurrence of VT [unadjusted odds ratio (OR) 4.507, 95% confidence interval (CI) 2.263-8.974, P < 0.001] and SA (OR 6.186, 95%CI 1.643-23.291, P = 0.007). After adjusting for age, sex, body mass index, and other confounding factors, the above differences were still statistically significant (VT: OR 4.808, 95%CI 2.254-10.253, P < 0.001; SA: OR 9.589, 95%CI 2.027-45.367, P = 0.004).NAFLD is associated with the occurrence of VT and SA in NSTEMI patients. It indicates that NAFLD might be a risk factor for malignant arrhythmias in post-NSTEMI patients.


Assuntos
Parada Cardíaca , Infarto do Miocárdio sem Supradesnível do Segmento ST , Hepatopatia Gordurosa não Alcoólica , Intervenção Coronária Percutânea , Infarto do Miocárdio com Supradesnível do Segmento ST , Taquicardia Ventricular , Complexos Ventriculares Prematuros , Parada Cardíaca/complicações , Humanos , Infarto do Miocárdio sem Supradesnível do Segmento ST/complicações , Hepatopatia Gordurosa não Alcoólica/complicações , Intervenção Coronária Percutânea/efeitos adversos , Fatores de Risco , Infarto do Miocárdio com Supradesnível do Segmento ST/complicações , Taquicardia Ventricular/complicações , Taquicardia Ventricular/etiologia , Complexos Ventriculares Prematuros/etiologia
4.
J Am Heart Assoc ; 10(21): e021895, 2021 11 02.
Artigo em Inglês | MEDLINE | ID: mdl-34713723

RESUMO

Background Exploring potential therapeutic target is of great significance for myocardial infarction (MI) and post-MI heart failure. Transcription factor Yin-Yang 1 (YY1) is an essential regulator of apoptosis and angiogenesis, but its role in MI is unclear. Methods and Results The expression of YY1 was assessed in the C57BL/6J mouse heart following MI. Overexpression or silencing of YY1 in the mouse heart was achieved by adeno-associated virus 9 injection. The survival, cardiac function, and scar size, as well as the apoptosis, angiogenesis, cardiac fibrosis, T helper 2 lymphocyte cytokine production, and macrophage polarization were assessed. The effects of YY1 on Akt phosphorylation and vascular endothelial growth factor production were also investigated. The expression of YY1 in heart was significantly stimulated by MI. The survival rate, cardiac function, scar size, and left ventricular volume of mice were improved by YY1 overexpression but worsened by YY1 silencing. YY1 alleviated cardiac apoptosis and fibrosis, promoted angiogenesis, T helper 2 cytokine production, and M2 macrophage polarization in the post-MI heart, it also enhanced the tube formation and migration ability of endothelial cells. Enhanced Akt phosphorylation, along with the increased vascular endothelial growth factor levels were observed in presence of YY1 overexpression. Conclusions YY1 ameliorates cardiac injury and remodeling after MI by repressing cardiomyocyte apoptosis and boosting angiogenesis, which might be ascribed to the enhancement of Akt phosphorylation and the subsequent vascular endothelial growth factor up-regulation. Increased T helper 2 cytokine production and M2 macrophage polarization may also be involved in YY1's cardioprotective effects. These findings supported YY1 as a potential target for therapeutic investigation of MI.


Assuntos
Traumatismos Cardíacos , Infarto do Miocárdio , Animais , Apoptose , Cicatriz , Citocinas , Modelos Animais de Doenças , Células Endoteliais , Camundongos , Camundongos Endogâmicos C57BL , Infarto do Miocárdio/prevenção & controle , Miócitos Cardíacos , Proteínas Proto-Oncogênicas c-akt , Fator A de Crescimento do Endotélio Vascular , Remodelação Ventricular , Yin-Yang
5.
Sci Rep ; 11(1): 15836, 2021 08 04.
Artigo em Inglês | MEDLINE | ID: mdl-34349170

RESUMO

Acute myocardial infarction (MI) is one of the leading causes of death in humans. Our previous studies showed that gastrin alleviated acute myocardial ischaemia-reperfusion injury. We hypothesize that gastrin might protect against heart injury after MI by promoting angiogenesis. An MI model was simulated by ligating the anterior descending coronary artery in adult male C57BL/6J mice. Gastrin was administered twice daily by intraperitoneal injection for 2 weeks after MI. We found that gastrin reduced mortality, improved myocardial function with reduced infarct size and promoted angiogenesis. Gastrin increased HIF-1α and VEGF expression. Downregulation of HIF-1α expression by siRNA reduced the proliferation, migration and tube formation of human umbilical vein endothelial cells. These results indicate that gastrin restores cardiac function after MI by promoting angiogenesis via the HIF-1α/VEGF pathway.


Assuntos
Gastrinas/farmacologia , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Infarto do Miocárdio/tratamento farmacológico , Neovascularização Patológica/prevenção & controle , Fator A de Crescimento do Endotélio Vascular/metabolismo , Animais , Proliferação de Células , Células Endoteliais da Veia Umbilical Humana/metabolismo , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Infarto do Miocárdio/patologia , Transdução de Sinais , Fator A de Crescimento do Endotélio Vascular/genética
6.
J Cardiovasc Transl Res ; 14(4): 610-618, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-32144627

RESUMO

Ischemia reperfusion (I/R)-induced arrhythmia is a serious complication in patients with cardiac infarction. Remodeling of connexin (Cx) 43, manifested as phosphorylation, contributes significantly to arrhythmogenesis. Integrin-linked kinase (ILK) attenuated ventricular remodeling and improved cardiac function in rats after myocardial infarction. We hypothesized that ILK, through Cx43 phosphorylation, would be protective against I/R-induced ventricular arrhythmias. Our study showed that I/R-induced ventricular arrhythmias were attenuated by an ILK agonist LPTP and worsened by the ILK inhibitor Cpd22. I/R disrupted Cx43 distribution, but it was partially normalized in the presence of LPTP. Compared with I/R, the phosphorylation of Akt was increased significantly after pretreatment with LPTP. The increase in phosphorylated Akt was physiologically significant because, in the presence of the Akt inhibitor MK2206, the protective effects of LPTP were blocked. This indicated that ILK activation prevented I/R-induced-ventricular arrhythmia, an effect potentially related to inhibition of Cx43 remodeling via Akt activation.


Assuntos
Antiarrítmicos/farmacologia , Conexina 43/metabolismo , Ativadores de Enzimas/farmacologia , Traumatismo por Reperfusão Miocárdica/tratamento farmacológico , Miócitos Cardíacos/efeitos dos fármacos , Proteínas Serina-Treonina Quinases/metabolismo , Taquicardia Ventricular/prevenção & controle , Fibrilação Ventricular/prevenção & controle , Animais , Modelos Animais de Doenças , Ativação Enzimática , Frequência Cardíaca/efeitos dos fármacos , Preparação de Coração Isolado , Masculino , Traumatismo por Reperfusão Miocárdica/enzimologia , Traumatismo por Reperfusão Miocárdica/patologia , Traumatismo por Reperfusão Miocárdica/fisiopatologia , Miócitos Cardíacos/enzimologia , Miócitos Cardíacos/patologia , Fosforilação , Proteínas Proto-Oncogênicas c-akt/metabolismo , Ratos Sprague-Dawley , Transdução de Sinais , Taquicardia Ventricular/enzimologia , Taquicardia Ventricular/patologia , Taquicardia Ventricular/fisiopatologia , Fibrilação Ventricular/enzimologia , Fibrilação Ventricular/patologia , Fibrilação Ventricular/fisiopatologia , Remodelação Ventricular/efeitos dos fármacos
7.
Circ Res ; 124(12): 1760-1777, 2019 06 07.
Artigo em Inglês | MEDLINE | ID: mdl-30982412

RESUMO

RATIONALE: PKA (Protein Kinase A) is a major mediator of ß-AR (ß-adrenergic) regulation of cardiac function, but other mediators have also been suggested. Reduced PKA basal activity and activation are linked to cardiac diseases. However, how complete loss of PKA activity impacts on cardiac physiology and if it causes cardiac dysfunction have never been determined. OBJECTIVES: We set to determine how the heart adapts to the loss of cardiomyocyte PKA activity and if it elicits cardiac abnormalities. METHODS AND RESULTS: (1) Cardiac PKA activity was almost completely inhibited by expressing a PKA inhibitor peptide in cardiomyocytes (cPKAi) in mice; (2) cPKAi reduced basal phosphorylation of 2 myofilament proteins (TnI [troponin I] and cardiac myosin binding protein C), and one longitudinal SR (sarcoplasmic reticulum) protein (PLB [phospholamban]) but not of the sarcolemmal proteins (Cav1.2 α1c and PLM [phospholemman]), dyadic protein RyR2, and nuclear protein CREB (cAMP response element binding protein) at their PKA phosphorylation sites; (3) cPKAi increased the expression of CaMKII (Ca2+/calmodulin-dependent kinase II), the Cav1.2 ß subunits and current, but decreased CaMKII phosphorylation and CaMKII-mediated phosphorylation of PLB and RyR2; (4) These changes resulted in significantly enhanced myofilament Ca2+ sensitivity, prolonged contraction, slowed relaxation but increased myocyte Ca2+ transient and contraction amplitudes; (5) Isoproterenol-induced PKA and CaMKII activation and their phosphorylation of proteins were prevented by cPKAi; (6) cPKAi abolished the increases of heart rate, and cardiac and myocyte contractility by a ß-AR agonist (isoproterenol), showing an important role of PKA and a minimal role of PKA-independent ß-AR signaling in acute cardiac regulation; (7) cPKAi mice have partial exercise capability probably by enhancing vascular constriction and ventricular filling during ß-AR stimulation; and (8) cPKAi mice did not show any cardiac functional or structural abnormalities during the 1-year study period. CONCLUSIONS: PKA activity suppression induces a unique Ca2+ handling phenotype, eliminates ß-AR regulation of heart rates and cardiac contractility but does not cause cardiac abnormalities.


Assuntos
Proteínas Quinases Dependentes de AMP Cíclico/antagonistas & inibidores , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Contração Miocárdica/fisiologia , Miócitos Cardíacos/metabolismo , Inibidores de Proteínas Quinases/farmacologia , Receptores Adrenérgicos beta/metabolismo , Agonistas Adrenérgicos beta/farmacologia , Sequência de Aminoácidos , Animais , Células Cultivadas , Proteínas Quinases Dependentes de AMP Cíclico/genética , Feminino , Masculino , Camundongos , Camundongos Transgênicos , Contração Miocárdica/efeitos dos fármacos , Miócitos Cardíacos/efeitos dos fármacos , Canal de Liberação de Cálcio do Receptor de Rianodina/metabolismo
8.
Acta Pharmacol Sin ; 40(1): 9-12, 2019 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-30002488

RESUMO

Myocardial infarction (MI) is one of the most serious health threats, resulting in huge physical and economic burdens worldwide. Wnt signaling pathways play an important role in developmental processes such as tissue patterning, cell differentiation and cell division. Appropriate regulation of the activities of Wnt signaling pathways is also important for heart development and healing in post-MI heart. Moreover, Wnt pathway inhibitors have been identified as novel antitumor drugs and applied in ongoing clinical trials. This research progress has generated increasing interests for investigating the effects of Wnt pathway inhibitors on MI healing. In this short review, we summarize the roles of Wnt signaling pathways in post-MI heart and the therapeutic effects of Wnt pathway inhibitors on MI, and discuss the underlying mechanisms of Wnt pathway inhibitors in cardiac repairing.


Assuntos
Infarto do Miocárdio/fisiopatologia , Via de Sinalização Wnt/efeitos dos fármacos , Animais , Cardiotônicos/uso terapêutico , Fibrose/fisiopatologia , Coração/fisiopatologia , Inflamação/fisiopatologia , Infarto do Miocárdio/tratamento farmacológico , Neovascularização Fisiológica/fisiologia , Via de Sinalização Wnt/fisiologia
9.
Biochim Biophys Acta Mol Basis Dis ; 1864(10): 3459-3467, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-30076960

RESUMO

WNT pathways are critically involved in the cardiac hypertrophy growth. Porcupine, an acyltransferase that specifically enables secretion of all WNT ligands, became a highly druggable target for inhibiting WNT pathways. Here we test if a novel small-molecule porcupine inhibitor CGX1321, which has entered human clinical trials as an anti-cancer agent, exerts an anti-hypertrophic effect. Transverse aortic constriction (TAC) was performed to induce cardiac hypertrophy on four-month-old male C57 mice. Cardiac function was measured with echocardiography. Histological analysis was performed to detect cardiomyocyte size and molecular expressions. CGX1321 was administrated daily for 4 weeks post TAC injury. As a result, CGX1321 improved cardiac function and animal survival of post-TAC mice. CGX1321 significantly reduced cardiomyocyte hypertrophy, cardiomyocyte apoptosis and fibrosis induced by TAC injury. CGX1321 significantly inhibited TAC induced nuclear translocation of ß-catenin and the elevation of Frizzled-2, cyclin-D1 and c-myc expression, indicating its inhibitory effect on canonical WNT pathway. Furthermore, CGX1321 inhibited TAC induced nuclear translocation of nuclear factor of activated T-cells and the elevation of phosphorylated c-Jun expression, suggesting its inhibitory function on non-canonical WNT pathway. We conclude that CGX1321 inhibits both canonical and non-canonical WNT pathways, and attenuates cardiac hypertrophy. Our findings support the porcupine inhibitors as a class of new drugs to be potentially used for treating patients with cardiac hypertrophy.


Assuntos
Aciltransferases/antagonistas & inibidores , Cardiomegalia/tratamento farmacológico , Inibidores Enzimáticos/administração & dosagem , Via de Sinalização Wnt/efeitos dos fármacos , Aciltransferases/genética , Animais , Apoptose/efeitos dos fármacos , Cardiomegalia/etiologia , Cardiomegalia/metabolismo , Modelos Animais de Doenças , Inibidores Enzimáticos/farmacologia , Masculino , Camundongos , Transporte Proteico , Proteínas Proto-Oncogênicas c-jun/metabolismo , Ratos
10.
J Am Heart Assoc ; 7(14)2018 07 12.
Artigo em Inglês | MEDLINE | ID: mdl-30005556

RESUMO

BACKGROUND: Ischemia/reperfusion injury (IRI) is one of the most predominant complications of ischemic heart disease. Gastrin has emerged as a regulator of cardiovascular function, playing a key protective role in hypoxia. Serum gastrin levels are increased in patients with myocardial infarction, but the pathophysiogical significance of this finding is unknown. The purpose of this study was to determine whether and how gastrin protects cardiac myocytes from IRI. METHODS AND RESULTS: Adult male Sprague-Dawley rats were used in the experiments. The hearts in living rats or isolated Langendorff-perfused rat hearts were subjected to ischemia followed by reperfusion to induce myocardial IRI. Gastrin, alone or with an antagonist, was administered before the induction of myocardial IRI. We found that gastrin improved myocardial function and reduced the expression of myocardial injury markers, infarct size, and cardiomyocyte apoptosis induced by IRI. Gastrin increased the phosphorylation levels of ERK1/2 (extracellular signal-regulated kinase 1/2), AKT (protein kinase B), and STAT3 (signal transducer and activator of transcription 3), indicating its ability to activate the RISK (reperfusion injury salvage kinase) and SAFE (survivor activating factor enhancement) pathways. The presence of inhibitors of ERK1/2, AKT, or STAT3 abrogated the gastrin-mediated protection. The protective effect of gastrin was via CCK2R (cholecystokinin 2 receptor) because the CCK2R blocker CI988 prevented the gastrin-mediated protection of the heart with IRI. Moreover, we found a negative correlation between serum levels of cardiac troponin I and gastrin in patients with unstable angina pectoris undergoing percutaneous coronary intervention, suggesting a protective effect of gastrin in human cardiomyocytes. CONCLUSIONS: These results indicate that gastrin can reduce myocardial IRI by activation of the RISK and SAFE pathways.


Assuntos
Gastrinas/farmacologia , Coração/efeitos dos fármacos , Hormônios/farmacologia , Traumatismo por Reperfusão Miocárdica/metabolismo , Miocárdio/metabolismo , Idoso , Angina Instável/sangue , Angina Instável/cirurgia , Animais , Apoptose/efeitos dos fármacos , Feminino , Gastrinas/sangue , Humanos , Preparação de Coração Isolado , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Masculino , Pessoa de Meia-Idade , Infarto do Miocárdio/metabolismo , Infarto do Miocárdio/patologia , Miocárdio/patologia , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/patologia , Intervenção Coronária Percutânea , Inibidores de Proteínas Quinases/farmacologia , Proteínas Proto-Oncogênicas c-akt/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-akt/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-akt/metabolismo , Ratos , Receptor de Colecistocinina B/antagonistas & inibidores , Receptor de Colecistocinina B/metabolismo , Fator de Transcrição STAT3/antagonistas & inibidores , Fator de Transcrição STAT3/efeitos dos fármacos , Fator de Transcrição STAT3/metabolismo , Transdução de Sinais , Troponina I/sangue
11.
Stem Cells Int ; 2018: 7045245, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30622568

RESUMO

Myocardial infarction (MI) is one of the leading causes of death worldwide. Mesenchymal stem cell (MSC) transplantation is considered a promising approach and has made significant progress in preclinical studies and clinical trials for treating MI. However, hurdles including poor survival, retention, homing, and differentiation capacity largely limit the therapeutic effect of transplanted MSCs. Many strategies such as preconditioning, genetic modification, cotransplantation with bioactive factors, and tissue engineering were developed to improve the survival and function of MSCs. On the other hand, optimizing the hostile transplantation microenvironment of the host myocardium is also of importance. Here, we review the modifications of MSCs as well as the host myocardium to improve the efficacy of MSC-based therapy against MI.

12.
Curr Stem Cell Res Ther ; 12(8): 624-636, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28969536

RESUMO

BACKGROUND: Despite optimal treatment, myocardial infarction (MI) remains a major cause of death worldwide. Stem cell transplantation, as a promising therapy hoping to improve myocardial function and generate new myocardium, has been intensely studied for treating MI. OBJECTIVE: To summarize the recent advance of stem cell transplantation for treating MI in clinical trials and preclinical studies. RESULTS: Several types of adult stem cells have been applied to clinical trials and exerted beneficial effects against MI. The therapeutic effects of these stem cells are varied, modest and mostly depend on paracrine function. Embryonic stem cells and induced pluripotent stem cells, as well as their derivative cells, bring a new dawn for cardiac regeneration although they have not been applied in patients due to safety concerns. Feasible solutions are required to stress the safety issue, and to improve the survival, engraftment, migration, differentiation and synchronization with recipient myocardium of transplanted stem cells. CONCLUSION: Stem cell transplantation for treating MI in clinical trials has achieved modest therapeutic effect. The hurdles limiting the stem cell function define the direction of further research.


Assuntos
Infarto do Miocárdio/terapia , Transplante de Células-Tronco/métodos , Animais , Humanos
13.
Biochim Biophys Acta Mol Basis Dis ; 1863(8): 2085-2092, 2017 08.
Artigo em Inglês | MEDLINE | ID: mdl-28249798

RESUMO

Mesenchymal stem cells (MSCs) exert therapeutic effect on treating acute myocardial infarction. Recent evidence showed that paracrine function rather than direct differentiation predominately contributes to the beneficial effects of MSCs, but how the paracrine factors function are not fully elucidated. In the present study, we tested if extracellular vesicles (EVs) secreted by MSC promotes angiogenesis in infracted heart via microRNAs. Immunostaining of CD31 and matrigel plug assay were performed to detect angiogenesis in a mouse myocardial infarction (MI) model. The cardiac function and structure was examined with echocardiographic analysis. Capillary-like tube formation, migration and proliferation of human umbilical vein endothelial cells (HUVECs) were determined. As a result, MSC-EVs significantly improved angiogenesis and cardiac function in post-MI heart. MSC-EVs increased the proliferation, migration and tube formation capacity of HUVECs. MicroRNA (miR)-210 was found to be enriched in MSC-EVs. The EVs collected from MSCs with miR-210 silence largely lost the pro-angiogenic effect both in-vitro and in-vivo. The miR-210 target gene Efna3, which plays a role in angiogenesis, was down-regulated by MSC-EVs treatment in HUVECs. In conclusion, MSC-EVs are sufficient to improve angiogenesis and exert therapeutic effect on MI, its pro- angiogenesis effect might be associated with a miR-210-Efna3 dependent mechanism. This article is part of a Special Issue entitled: Genetic and epigenetic control of heart failure - edited by Jun Ren & Megan Yingmei Zhang.


Assuntos
Micropartículas Derivadas de Células/transplante , Células-Tronco Mesenquimais/metabolismo , MicroRNAs/metabolismo , Infarto do Miocárdio/terapia , Neovascularização Fisiológica , Animais , Micropartículas Derivadas de Células/metabolismo , Micropartículas Derivadas de Células/patologia , Células Endoteliais da Veia Umbilical Humana/metabolismo , Células Endoteliais da Veia Umbilical Humana/patologia , Humanos , Células-Tronco Mesenquimais/patologia , Camundongos , MicroRNAs/genética , Infarto do Miocárdio/genética , Infarto do Miocárdio/metabolismo , Infarto do Miocárdio/patologia
14.
Stem Cell Res Ther ; 8(1): 17, 2017 01 28.
Artigo em Inglês | MEDLINE | ID: mdl-28129786

RESUMO

BACKGROUND: Transplantation of cardiosphere-derived cells (CDCs) has been shown to exert a therapeutic effect in patients with myocardial infarction (MI). However, poor survival of transplanted CDCs limits their beneficial effect. Metformin (MET) activates AMP-activated protein kinase (AMPK) which is associated with cell survival. The aim of this study is to determine whether MET improves CDC survival in the transplantation microenvironment and enhances the therapeutic effect of CDC transplantation against MI. METHODS: CDCs were isolated and expanded from transgenic ß-actin-GFP mice. CDCs were pretreated with MET and intramyocardially injected into wild-type C57 mouse heart with MI injury. The survival of CDCs was quantified, and the infarct size and cardiac function of treated hearts were evaluated. RESULTS: CDC transplantation modestly reduced infarct size and improved cardiac function in the post-MI heart, which was further improved by MET treatment. MET pretreatment significantly increased the survival of CDCs transplanted into the myocardium. MET also reduced CDC apoptosis induced by oxidative stress in vitro. The anti-apoptotic effect of MET was blocked by the AMPK inhibitor compound C. MET increased AMPK phosphorylation and upregulated endothelial nitric oxide synthase (eNOS) in CDCs under oxidative stress, which might be associated with the anti-apoptotic effect of MET. CONCLUSIONS: MET improves the survival of transplanted CDCs in the myocardium, thereby enhancing their therapeutic effect against MI injury. The pro-survival function of MET on CDCs might be associated with an AMPK-eNOS-dependent mechanism.


Assuntos
Proteínas Quinases Ativadas por AMP/genética , Hipoglicemiantes/farmacologia , Metformina/farmacologia , Infarto do Miocárdio/terapia , Esferoides Celulares/transplante , Proteínas Quinases Ativadas por AMP/metabolismo , Animais , Proliferação de Células , Sobrevivência Celular/efeitos dos fármacos , Fibroblastos/citologia , Fibroblastos/metabolismo , Regulação da Expressão Gênica , Genes Reporter , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Humanos , Injeções Intralesionais , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Infarto do Miocárdio/genética , Infarto do Miocárdio/metabolismo , Infarto do Miocárdio/patologia , Miocárdio/metabolismo , Miocárdio/patologia , Miócitos Cardíacos/citologia , Miócitos Cardíacos/metabolismo , Óxido Nítrico Sintase Tipo III/genética , Óxido Nítrico Sintase Tipo III/metabolismo , Fosforilação/efeitos dos fármacos , Pirazóis/farmacologia , Pirimidinas/farmacologia , Esferoides Celulares/citologia , Esferoides Celulares/metabolismo
15.
Biochem Biophys Res Commun ; 474(3): 599-605, 2016 06 03.
Artigo em Inglês | MEDLINE | ID: mdl-27018249

RESUMO

Tirofiban, a glycoprotein IIb/IIIa inhibitor, is an antiplatelet drug extensively used in patients with acute coronary syndrome (ACS) and exerts an therapeutic effect on no-reflow phenomenon during percutaneous coronary intervention (PCI). Previous studies elucidated the vasodilation caused by tirofiban in the peripheral artery. However, whether tirofiban exerts a vasodilator effect on the coronary artery is unclear. Our present study found that tirofiban induced endothelium-dependent vasodilation in a concentration- and time-dependent manner in the isolated rat coronary artery pre-constricted by 5-hydroxytryptamine (5-HT). Further study showed that incubation of human umbilical venous endothelial cells (HUVECs) with tirofiban increased NO production, which was ascribed to the increased eNOS phosphorylation. This was confirmed by the loss of the vasorelaxant effect of tirofiban in the presence of l-NAME (eNOS inhibitor) and L-NMMA (NOS inhibitor) but not SMT (iNOS inhibitor) on isolated rat coronary arteries. The vasorelaxation was also blocked by the PI3K inhibitors, wortmannin and LY294002, as well as the Akt inhibitor SH-5, indicating the role of PI3K and Akt in tirofiban-mediated vasodilation. Moreover, further study showed that soluble guanylyl cyclase (sGC) inhibitor ODQ, or blockers of potassium channel (big-conductance calcium-activated potassium channel) blocked tirofiban-induced vasodilation of the coronary artery. These findings suggest that tirofiban induces vasorelaxation via an endothelium-dependent NO-cGMP signaling through the activation of the Akt/eNOS/sGC pathway.


Assuntos
Vasos Coronários/fisiologia , GMP Cíclico/metabolismo , Endotélio Vascular/metabolismo , Óxido Nítrico/metabolismo , Tirosina/análogos & derivados , Vasodilatação/fisiologia , Animais , Circulação Coronária/efeitos dos fármacos , Circulação Coronária/fisiologia , Vasos Coronários/efeitos dos fármacos , Relação Dose-Resposta a Droga , Endotélio Vascular/efeitos dos fármacos , Técnicas In Vitro , Masculino , Óxido Nítrico Sintase Tipo III/metabolismo , Fosfatidilinositol 3-Quinase/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Ratos , Ratos Wistar , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Tirofibana , Tirosina/administração & dosagem , Vasodilatação/efeitos dos fármacos , Vasodilatadores/administração & dosagem
16.
Stem Cells Int ; 2016: 9682757, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26681958

RESUMO

Mesenchymal stem cell (MSC) is an intensely studied stem cell type applied for cardiac repair. For decades, the preclinical researches on animal model and clinical trials have suggested that MSC transplantation exerts therapeutic effect on ischemic heart disease. However, there remain major limitations to be overcome, one of which is the very low survival rate after transplantation in heart tissue. Various strategies have been tried to improve the MSC survival, and many of them showed promising results. In this review, we analyzed the studies in recent years to summarize the methods, effects, and mechanisms of the new strategies to address this question.

17.
Hypertens Res ; 38(12): 807-12, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26178154

RESUMO

Proliferation of vascular smooth muscle cells (VSMCs) is thought to have a key role in the development of atherosclerotic lesions. Neuropeptide Y (NPY), norepinephrine and dopamine are sympathetic neurotransmitters. NPY has been particularly shown to stimulate proliferation of VSMCs. NPY, norepinephrine and dopamine are all sympathetic transmitters. In our previous study, we found that in the presence of the dopamine receptor, the α1-adrenergic receptor-mediated VSMC proliferation is reduced. We hypothesize that the activation of the D1-like receptor might inhibit the NPY-mediated VSMC proliferation. In our present study, we found that NPY, mainly via the Y1 receptor, increased VSMC proliferation. This was determined by [(3)H]-thymidine incorporation, in a concentration (10(-11) to 10(-8) M)-dependent manner. In the presence of the D1-like receptor agonist, fenoldopam (10(-12) to 10(-5) M), the stimulatory effect of NPY on VSMC proliferation was reduced. The involvement of the D1-like receptor was confirmed when the inhibitory effect of fenoldopam was reversed in the presence of the D1-like receptor antagonist SCH-23390 (10(-8) M). Moreover, the inhibitory effect of fenoldopam on NPY-mediated VSMC proliferation was also blocked in the presence of the PKA inhibitor 14-22 (10(-6) M). Protein kinase A activator 8-(4-chlorophenylthio) adenosine-3,5-cyclic monophosphorothioate, Sp-isomer sodium salt (10(-6) M) could simulate the stimulatory effect of fenoldopam. It indicated that the inhibitory effect of D1-like receptors on NPY-mediated VSMC proliferation may have an important role in the regulation of blood pressure or prevention of atherosclerosis.


Assuntos
Músculo Liso Vascular/efeitos dos fármacos , Miócitos de Músculo Liso/efeitos dos fármacos , Neuropeptídeo Y/farmacologia , Receptores de Dopamina D1/fisiologia , Animais , Aterosclerose/etiologia , Proliferação de Células/efeitos dos fármacos , Proteínas Quinases Dependentes de AMP Cíclico/fisiologia , Fenoldopam/farmacologia , Masculino , Músculo Liso Vascular/citologia , Miócitos de Músculo Liso/fisiologia , Neointima , Ratos , Ratos Sprague-Dawley
18.
Hypertens Res ; 38(9): 588-96, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25832920

RESUMO

Angiotensin (Ang) II has an important role in the vascular smooth muscle cell (VSMC) proliferation and migration and subsequently in the development of vascular diseases, whereas dopamine has the opposite effect. Previous studies have shown an interaction between dopamine and AT(1) receptors in the kidney. The dopamine D(4) receptor is expressed in arteries and has an inhibitory effect on VSMC proliferation. We hypothesized that the D(4) receptor, through its interaction with the AT(1a) receptor, may have an inhibitory effect on Ang II-mediated VSMC proliferation and migration, which could have a pivotal role in hypertension-induced vascular remodeling. In the current study, we found that Ang II markedly induced the proliferation and migration of A10 cells, which was inhibited by the D(4) receptor agonist PD168077. The activation of the D(4) receptor by PD168077 inhibited AT(1a) receptor expression in a concentration- and time-dependent manner. These effects were attenuated by silencing the D(4) receptor with a D(4) receptor-targeting small interfering RNA. The D(4) receptor-mediated inhibition of AT(1) receptor function involved protein kinase A (PKA). The activation of the D(4) receptor by PD168077 increased PKA activity in A10 cells, and the presence of a PKA inhibitor (PKA inhibitor 14-22, 10(-7) mol l(-1) per 24 h) blocked the inhibitory effect of the D(4) receptor on AT(1) receptor expression and function. The inhibitory effect of the D(4) receptor on AT(1) receptor expression and function was preserved in VSMCs (primary culture) from spontaneously hypertensive rats relative to VSMCs from Wistar-Kyoto rats. In conclusion, our data provide insight into the regulatory role of the D(4) receptor on AT(1a) receptor expression and function in VSMCs and suggest that targeting the action of the D(4) receptor may represent an effective therapeutic approach for the treatment of cardiovascular diseases.


Assuntos
Agonistas de Dopamina/farmacologia , Músculo Liso Vascular/efeitos dos fármacos , Miócitos de Músculo Liso/efeitos dos fármacos , Receptor Tipo 1 de Angiotensina/biossíntese , Receptores de Dopamina D4/agonistas , Animais , Benzamidas/farmacologia , Linhagem Celular , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Relação Dose-Resposta a Droga , Regulação para Baixo , Inativação Gênica , Piperazinas/farmacologia , RNA Interferente Pequeno , Ratos , Ratos Endogâmicos SHR , Ratos Endogâmicos WKY , Receptor Tipo 1 de Angiotensina/efeitos dos fármacos
19.
J Thromb Thrombolysis ; 37(2): 102-6, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-23584598

RESUMO

P2Y12 receptor antagonists have become cornerstone pharmacological agents in antiplatelet therapy for patients with acute coronary syndrome or undergoing percutaneous coronary intervention. While clopidogrel remains in extensive use in clinical practice, it cannot meet the needs in many clinical conditions because of its pharmacological limitations. In recent years, newly developed P2Y12 antagonists, such as prasugrel and ticagrelor, have proven to be of higher efficacy and less resistance. With the introduction of these new medicines, the current antiplatelet strategy is undergoing a transitional period. Insufficient platelet inhibition (high platelet reactivity) leads to an increased risk of ischemic events whereas exceeding platelet inhibition can lead to an increased risk of bleeding. This review discusses the pharmacological features, benefits, risks and cost-effectiveness of different P2Y12 antagonists in various clinical settings. A balance between these factors will determine the choice of P2Y12 antagonists for personalized antiplatelet therapy. We conclude that it is a promising option to apply the new drugs, due to their superior therapeutic performance versus that of clopidogrel in the long run.


Assuntos
Síndrome Coronariana Aguda/tratamento farmacológico , Adenosina/análogos & derivados , Piperazinas/uso terapêutico , Antagonistas do Receptor Purinérgico P2Y/uso terapêutico , Tiofenos/uso terapêutico , Adenosina/efeitos adversos , Adenosina/uso terapêutico , Clopidogrel , Hemorragia/induzido quimicamente , Humanos , Piperazinas/efeitos adversos , Cloridrato de Prasugrel , Antagonistas do Receptor Purinérgico P2Y/efeitos adversos , Fatores de Risco , Tiofenos/efeitos adversos , Ticagrelor , Ticlopidina/efeitos adversos , Ticlopidina/análogos & derivados , Ticlopidina/uso terapêutico
20.
Clin Sci (Lond) ; 125(7): 319-27, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23746375

RESUMO

Stem cell therapy has emerged as a promising strategy for cardiac and vascular repair. The ultimate goal is to rebuild functional myocardium by transplanting exogenous stem cells or by activating native stem cells to induce endogenous repair. CS/PCs (cardiac stem/progenitor cells) are one type of adult stem cell with the potential to differentiate into cardiac lineages (cardiomyocytes, smooth muscle cells and endothelial cells). iPSCs (induced pluripotent stem cells) also have the capacity to differentiate into necessary cells to rebuild injured cardiac tissue. Both types of stem cells have brought promise for cardiac repair. The present review summarizes recent advances in cardiac cell therapy based on these two cell sources and discusses the advantages and limitations of each candidate. We conclude that, although both types of stem cells can be considered for autologous transplantation with promising outcomes in animal models, CS/PCs have advanced more in their clinical application because iPSCs and their derivatives possess inherent obstacles for clinical use. Further studies are needed to move cell therapy forward for the treatment of heart disease.


Assuntos
Células-Tronco Pluripotentes Induzidas/transplante , Isquemia Miocárdica/terapia , Transplante de Células-Tronco/métodos , Animais , Diferenciação Celular , Modelos Animais de Doenças , Humanos , Miócitos Cardíacos/transplante
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA