Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Biochem Biophys Res Commun ; 690: 149272, 2024 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-37992523

RESUMO

Cardiomyocytes (CMs) have little regenerative capacity. After myocardial infarction (MI), scar formation and myocardial remodeling proceed in the infarct and non-infarct areas, respectively, leading to heart failure (HF). Prolonged activation of cardiac fibroblasts (CFs) and inflammatory cells may contribute to this process; however, therapies targeting these cell types remain lacking. Cardiac reprogramming converts CFs into induced CMs, reduces fibrosis, and improves cardiac function in chronic MI through the overexpression of Mef2c/Gata4/Tbx5/Hand2 (MGTH). However, whether cardiac reprogramming reduces inflammation in infarcted hearts remains unclear. Moreover, the mechanism through which MGTH overexpression in CFs affects inflammatory cells remains unknown. Here, we showed that inflammation persists in the myocardium until three months after MI, which can be reversed with cardiac reprogramming. Single-cell RNA sequencing demonstrated that CFs expressed pro-inflammatory genes and exhibited strong intercellular communication with inflammatory cells, including macrophages, in chronic MI. Cardiac reprogramming suppressed the inflammatory profiles of CFs and reduced the relative ratios and pro-inflammatory signatures of cardiac macrophages. Moreover, fluorescence-activated cell sorting analysis (FACS) revealed that cardiac reprogramming reduced the number of chemokine receptor type 2 (CCR2)-positive inflammatory macrophages in the non-infarct areas in chronic MI, thereby restoring myocardial remodeling. Thus, cardiac reprogramming reduced the number of inflammatory macrophages to exacerbate cardiac function after MI.


Assuntos
Infarto do Miocárdio , Humanos , Infarto do Miocárdio/metabolismo , Miocárdio/metabolismo , Miócitos Cardíacos/metabolismo , Macrófagos/metabolismo , Inflamação/metabolismo , Fibroblastos/metabolismo
2.
Arterioscler Thromb Vasc Biol ; 43(9): 1668-1683, 2023 09.
Artigo em Inglês | MEDLINE | ID: mdl-37534464

RESUMO

BACKGROUND: The mechanisms underlying pulmonary hypertension (PH) remain largely unknown; further, why advanced vascular remodeling preferentially occurs in arterioles is yet to be answered. VEGF (vascular endothelial growth factor) regulates angiogenesis through Flk1 (fetal liver kinase 1) and Flt1 (fms-like tyrosine kinase 1) on endothelial cells (ECs), which may be related to PH pathogenesis. However, spatiotemporal expression patterns of Flk1 and Flt1 in the pulmonary vascular system and the role of endothelial Flk1 in PH development remain poorly understood. METHODS: We analyzed multiple reporter mice, including Flk1-GFP (green fluorescent protein) bacterial artificial chromosome transgenic (Tg), Flt1-DsRed bacterial artificial chromosome Tg, and Flk1-GFP/Flt1-DsRed double Tg mice, to determine the spatiotemporal expression of Flk1 and Flt1 in hypoxia-induced PH. We also used Cdh5CreERT2/Flk1f/f/Tomato (Flk1-KO [knockout]) mice to induce EC-specific Flk1 deletion and lineage tracing in chronic hypoxia. RESULTS: Flk1 was specifically expressed in the ECs of small pulmonary vessels, including arterioles. Conversely, Flt1 was more broadly expressed in the ECs of large- to small-sized vessels in adult mouse lungs. Intriguingly, Flk1+ ECs were transiently increased in hypoxia with proliferation, whereas Flt1 expression was unchanged. Flk1-KO mice did not exhibit pulmonary vascular remodeling nor PH in normoxia; however, the arteriolar ECs changed to a cuboidal shape with protrusion. In hypoxia, Flk1 deletion exacerbated EC dysfunction and reduced their number via apoptosis. Additionally, Flk1 deletion promoted medial thickening and neointimal formation in arterioles and worsened PH. Mechanistically, lineage tracing revealed that neointimal cells were derived from Flk1-KO ECs. Moreover, RNA sequencing in pulmonary ECs demonstrated that Flk1 deletion and hypoxia synergistically activated multiple pathways, including cell cycle, senescence/apoptosis, and cytokine/growth factor, concomitant with suppression of cell adhesion and angiogenesis, to promote vascular remodeling. CONCLUSIONS: Flk1 and Flt1 were differentially expressed in pulmonary ECs. Flk1 deficiency and hypoxia jointly dysregulated arteriolar ECs to promote vascular remodeling. Thus, dysfunction of Flk1+ ECs may contribute to the pathogenesis of advanced vascular remodeling in pulmonary arterioles.


Assuntos
Hipertensão Pulmonar , Remodelação Vascular , Animais , Camundongos , Células Endoteliais/metabolismo , Proteínas de Fluorescência Verde/metabolismo , Hipertensão Pulmonar/genética , Hipertensão Pulmonar/metabolismo , Hipóxia/complicações , Hipóxia/genética , Hipóxia/metabolismo , Camundongos Knockout , Camundongos Transgênicos , Fator A de Crescimento do Endotélio Vascular/metabolismo
3.
Semin Cell Dev Biol ; 122: 21-27, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-34210577

RESUMO

Cardiovascular diseases are a common cause of death worldwide. Adult cardiomyocytes have limited regenerative capacity after injury, and there is growing interest in cardiac regeneration as a new therapeutic strategy. There are several limitations of induced pluripotent stem cell-based transplantation therapy with respect to efficiency and risks of tumorigenesis. Direct reprogramming enables the conversion of terminally differentiated cells into target cell types using defined factors. In most cardiac diseases, activated fibroblasts proliferate in the damaged heart and contribute to the progression of heart failure. In vivo cardiac reprogramming, in which resident cardiac fibroblasts are converted into cardiomyocytes in situ, is expected to become a new cardiac regenerative therapy. Indeed, we and other groups have demonstrated that in vivo reprogramming improves cardiac function and reduces fibrosis after myocardial infarction. In this review, we summarize recent discoveries and developments related to in vivo reprogramming. In addition, issues that need to be resolved for clinical application are described.


Assuntos
Reprogramação Celular/fisiologia , Cardiopatias/terapia , Miócitos Cardíacos/metabolismo , Medicina Regenerativa/métodos , Animais , Humanos , Camundongos
4.
Regen Ther ; 11: 95-100, 2019 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-31304202

RESUMO

Cardiovascular disease is the leading cause of death globally. Cardiomyocytes (CMs) have poor regenerative capacity, and pharmacological therapies have limited efficacy in severe heart failure. Currently, there are several promising strategies for cardiac regeneration. The most promising approach to remuscularize failing hearts is cell transplantation therapy using newly generated CMs from exogenous sources, such as pluripotent stem cells. Alternatively, approaches to generate new CMs from endogenous cell sources in situ may also repair the injured heart and improve cardiac function. Direct cardiac reprogramming has emerged as a novel therapeutic approach to regenerate injured hearts by directly converting endogenous cardiac fibroblasts into CM-like cells. Through cell transplantation and direct cardiac reprogramming, new CMs can be generated and scar tissue reduced to improve cardiac function; therefore, cardiac regeneration may serve as a powerful strategy for treatment of severe heart failure. While substantial progress has been made in these two strategies for cardiac regeneration over the past several years, challenges remain for clinical translation. This review provide an overview of previous reports and current challenges in this field.

5.
Biochem Biophys Res Commun ; 513(4): 1041-1047, 2019 06 11.
Artigo em Inglês | MEDLINE | ID: mdl-31010673

RESUMO

Cardiovascular disease is a leading cause of death worldwide. Mammalian cardiomyocytes (CMs) proliferate during embryonic development, whereas they largely lose their regenerative capacity after birth. Defined factors expressed in cardiac progenitors or embryonic CMs may activate the cell cycle and induce CM proliferation in postnatal and adult hearts. Here, we report that the overexpression of Tbx6, enriched in the cardiac mesoderm (progenitor cells), induces CM proliferation in postnatal and adult mouse hearts. By screening 24 factors enriched in cardiac progenitors or embryonic CMs, we found that only Tbx6 could induce CM proliferation in primary cultured postnatal rat CMs. Intriguingly, it did not induce the proliferation of cardiac fibroblasts. We next generated a recombinant adeno-associated virus serotype 9 vector encoding Tbx6 (AAV9-Tbx6) for transduction into mouse CMs in vivo. The subcutaneous injection of AAV9-Tbx6 into neonatal mice induced CM proliferation in postnatal and adult mouse hearts. Mechanistically, Tbx6 overexpression upregulated multiple cell cycle activators including Aurkb, Mki67, Ccna1, and Ccnb2 and suppressed the tumor suppressor Rb1. Thus, Tbx6 promotes CM proliferation in postnatal and adult mouse hearts by modifying the expression of cell cycle regulators.


Assuntos
Proliferação de Células/efeitos dos fármacos , Miocárdio/citologia , Miócitos Cardíacos/citologia , Proteínas com Domínio T/fisiologia , Adenoviridae/genética , Animais , Animais Recém-Nascidos , Proteínas de Ciclo Celular/efeitos dos fármacos , Células Cultivadas , Ciclinas/efeitos dos fármacos , Vetores Genéticos/administração & dosagem , Coração , Camundongos , Ratos , Regeneração , Proteínas com Domínio T/genética , Proteínas com Domínio T/metabolismo , Proteínas com Domínio T/farmacologia
6.
Nat Commun ; 10(1): 674, 2019 02 20.
Artigo em Inglês | MEDLINE | ID: mdl-30787297

RESUMO

Direct cardiac reprogramming from fibroblasts can be a promising approach for disease modeling, drug screening, and cardiac regeneration in pediatric and adult patients. However, postnatal and adult fibroblasts are less efficient for reprogramming compared with embryonic fibroblasts, and barriers to cardiac reprogramming associated with aging remain undetermined. In this study, we screened 8400 chemical compounds and found that diclofenac sodium (diclofenac), a non-steroidal anti-inflammatory drug, greatly enhanced cardiac reprogramming in combination with Gata4, Mef2c, and Tbx5 (GMT) or GMT plus Hand2. Intriguingly, diclofenac promoted cardiac reprogramming in mouse postnatal and adult tail-tip fibroblasts (TTFs), but not in mouse embryonic fibroblasts (MEFs). Mechanistically, diclofenac enhanced cardiac reprogramming by inhibiting cyclooxygenase-2, prostaglandin E2/prostaglandin E receptor 4, cyclic AMP/protein kinase A, and interleukin 1ß signaling and by silencing inflammatory and fibroblast programs, which were activated in postnatal and adult TTFs. Thus, anti-inflammation represents a new target for cardiac reprogramming associated with aging.


Assuntos
Reprogramação Celular/efeitos dos fármacos , Ciclo-Oxigenase 2/farmacologia , Miócitos Cardíacos/efeitos dos fármacos , Receptores de Prostaglandina E Subtipo EP4/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Animais , Anti-Inflamatórios não Esteroides/farmacologia , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Diferenciação Celular/efeitos dos fármacos , AMP Cíclico , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Ciclo-Oxigenase 2/efeitos dos fármacos , Diclofenaco/farmacologia , Dinoprostona , Fibroblastos , Fator de Transcrição GATA4/metabolismo , Humanos , Inflamação , Interleucina-1beta , Fatores de Transcrição MEF2/metabolismo , Camundongos , Camundongos Transgênicos , Proteínas com Domínio T/metabolismo
7.
J Cardiol ; 73(2): 97-101, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30420106

RESUMO

Cardiac muscle has limited proliferative capacity, and regenerative therapies are highly in demand as a new treatment strategy. Pharmacological and non-pharmacological therapies have been developed, but these medical therapies have limited effects to cure patients with severe heart failure. Moreover, heart transplantation is limited due to the low number of donor organs. Thus, heart regeneration holds great potential to offer innovative therapy to treat heart failure patients. Currently, there are several strategies for heart regeneration. Transplantation of somatic stem cells was safe and modestly improved cardiac function after myocardial infarction mainly through paracrine mechanisms. Alternatively, new cardiomyocytes could be generated from induced pluripotent stem cells (iPSCs) to transplant into injured hearts. However, several issues remain to be resolved prior to using iPSC-derived cardiomyocytes, such as a potential risk of tumorigenesis and poor survival of transplanted cells in the injured heart. More recently, direct cardiac reprogramming has emerged as a novel technology to regenerate damaged myocardium by directly converting endogenous cardiac fibroblasts into induced cardiomyocyte-like cells to restore cardiac function. Following our first report of cardiac reprogramming, an improvement in cardiac reprogramming efficiency, in vivo direct cardiac reprogramming, and cardiac reprogramming in human cells were reported by many investigators. While these previous studies have advanced regenerative research, many challenges remain. Here, we review the current status of cardiac regenerative technology, a great hope to treat cardiovascular diseases.


Assuntos
Regeneração Tecidual Guiada/métodos , Insuficiência Cardíaca/terapia , Coração/fisiopatologia , Animais , Diferenciação Celular , Fibroblastos/transplante , Insuficiência Cardíaca/fisiopatologia , Humanos , Células-Tronco Pluripotentes Induzidas/fisiologia , Infarto do Miocárdio/terapia , Miocárdio/patologia , Miócitos Cardíacos/transplante , Regeneração , Transplante de Células-Tronco/métodos
8.
Int J Mol Sci ; 19(9)2018 Sep 05.
Artigo em Inglês | MEDLINE | ID: mdl-30189626

RESUMO

Cardiac diseases are among the most common causes of death globally. Cardiac muscle has limited proliferative capacity, and regenerative therapies are highly in demand as a new treatment strategy. Although pluripotent reprogramming has been developed, it has obstacles, such as a potential risk of tumor formation, poor survival of the transplanted cells, and high cost. We previously reported that fibroblasts can be directly reprogrammed to cardiomyocytes by overexpressing a combination of three cardiac-specific transcription factors (Gata4, Mef2c, Tbx5 (together, GMT)). We and other groups have promoted cardiac reprogramming by the addition of certain miRNAs, cytokines, and epigenetic factors, and unraveled new molecular mechanisms of cardiac reprogramming. More recently, we discovered that Sendai virus (SeV) vector expressing GMT could efficiently and rapidly reprogram fibroblasts into integration-free cardiomyocytes in vitro via robust transgene expression. Gene delivery of SeV-GMT also improves cardiac function and reduces fibrosis after myocardial infarction in mice. Through direct cardiac reprogramming, new cardiomyocytes can be generated and scar tissue reduced to restore cardiac function, and, thus, direct cardiac reprogramming may serve as a powerful strategy for cardiac regeneration. Here, we provide an overview of the previous reports and current challenges in this field.


Assuntos
Reprogramação Celular , Coração/fisiologia , Miócitos Cardíacos/metabolismo , Regeneração , Animais , Diferenciação Celular , Epigênese Genética , Fibroblastos/metabolismo , Fator de Transcrição GATA4/genética , Fator de Transcrição GATA4/metabolismo , Regulação da Expressão Gênica , Humanos , Fatores de Transcrição MEF2/genética , Fatores de Transcrição MEF2/metabolismo , Proteínas com Domínio T/genética , Proteínas com Domínio T/metabolismo
9.
J Am Heart Assoc ; 7(1)2018 01 07.
Artigo em Inglês | MEDLINE | ID: mdl-29307865

RESUMO

BACKGROUND: It has never been possible to immediately evaluate heart rate variability (HRV) during exercise. We aimed to visualize the real-time changes in the power spectrum of HRV during exercise and to investigate its relationship to the ventilatory threshold (VT). METHODS AND RESULTS: Thirty healthy subjects (29.1±5.7 years of age) and 35 consecutive patients (59.0±13.2 years of age) with myocardial infarctions underwent cardiopulmonary exercise tests with an RAMP protocol ergometer. The HRV was continuously assessed with power spectral analyses using the maximum entropy method and projected on a screen without delay. During exercise, a significant decrease in the high frequency (HF) was followed by a drastic shift in the power spectrum of the HRV with a periodic augmentation in the low frequency/HF (L/H) and steady low HF. When the HRV threshold (HRVT) was defined as conversion from a predominant high frequency (HF) to a predominant low frequency/HF (L/H), the VO2 at the HRVT (HRVT-VO2) was substantially correlated with the VO2 at the lactate threshold and VT) in the healthy subjects (r=0.853 and 0.921, respectively). The mean difference between each threshold (0.65 mL/kg per minute for lactate threshold and HRVT, 0.53 mL/kg per minute for VT and HRVT) was nonsignificant (P>0.05). Furthermore, the HRVT-VO2 was also correlated with the VT-VO2 in these myocardial infarction patients (r=0.867), and the mean difference was -0.72 mL/kg per minute and was nonsignificant (P>0.05). CONCLUSIONS: A HRV analysis with our method enabled real-time visualization of the changes in the power spectrum during exercise. This can provide additional information for detecting the VT.


Assuntos
Limiar Anaeróbio , Aptidão Cardiorrespiratória , Eletrocardiografia/métodos , Teste de Esforço/métodos , Tolerância ao Exercício , Frequência Cardíaca , Pulmão/fisiopatologia , Infarto do Miocárdio/diagnóstico , Processamento de Sinais Assistido por Computador , Adulto , Idoso , Ciclismo , Biomarcadores/sangue , Estudos de Casos e Controles , Feminino , Humanos , Ácido Láctico/sangue , Masculino , Pessoa de Meia-Idade , Infarto do Miocárdio/sangue , Infarto do Miocárdio/fisiopatologia , Valor Preditivo dos Testes , Reprodutibilidade dos Testes , Mecânica Respiratória , Fatores de Tempo
10.
Biochem Biophys Res Commun ; 495(1): 884-891, 2018 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-29158084

RESUMO

The coronary vascular system is critical for myocardial growth and cardiomyocyte survival. However, the molecular mechanism regulating coronary angiogenesis remains elusive. Vascular endothelial growth factor (VEGF) regulates angiogenesis by binding to the specific receptors Flk1 and Flt1, which results in different functions. Despite the importance of Flk1 and Flt1, their expression in the coronary vasculature remains largely unknown due to the lack of appropriate antibodies for immunostaining. Here, we analyzed multiple reporter mice including Flk1-GFP BAC transgenic (Tg), Flk1-LacZ knock-in, Flt1-DsRed BAC Tg, and Flk1-GFP/Flt1-DsRed double Tg animals to determine expression patterns in mouse hearts during cardiac growth and after myocardial infarction (MI). We found that Flk1 was expressed in endothelial cells (ECs) with a pattern of epicardial-to-endocardial transmural gradients in the neonatal mouse ventricle, which was downregulated in adult coronary vessels with development. In contrast, Flt1 was homogeneously expressed in the ECs of neonatal mouse hearts and expression was maintained until adulthood. After MI, expression of both Flk1 and Flt1 was induced in the regenerating coronary vessels at day 7. Intriguingly, Flk1 expression was downregulated thereafter, whereas Flt1 expression was maintained in the newly formed coronary vessels until 30 days post-MI, recapitulating their expression kinetics during development. This is the first report demonstrating the spatiotemporal expression patterns of Flk1 and Flt1 in the coronary vascular system during development and after MI; thus, this study suggests that these factors have distinct and important functions in coronary angiogenesis.


Assuntos
Envelhecimento/metabolismo , Vasos Coronários/metabolismo , Infarto do Miocárdio/metabolismo , Miocárdio/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo , Receptor 1 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Animais , Vasos Coronários/crescimento & desenvolvimento , Progressão da Doença , Regulação da Expressão Gênica no Desenvolvimento , Camundongos , Neovascularização Fisiológica/fisiologia
11.
Stem Cell Reports ; 5(6): 1128-1142, 2015 Dec 08.
Artigo em Inglês | MEDLINE | ID: mdl-26626177

RESUMO

Fibroblasts can be directly reprogrammed into cardiomyocyte-like cells (iCMs) by overexpression of cardiac transcription factors, including Gata4, Mef2c, and Tbx5; however, this process is inefficient under serum-based culture conditions, in which conversion of partially reprogrammed cells into fully reprogrammed functional iCMs has been a major hurdle. Here, we report that a combination of fibroblast growth factor (FGF) 2, FGF10, and vascular endothelial growth factor (VEGF), termed FFV, promoted cardiac reprogramming under defined serum-free conditions, increasing spontaneously beating iCMs by 100-fold compared with those under conventional serum-based conditions. Mechanistically, FFV activated multiple cardiac transcriptional regulators and converted partially reprogrammed cells into functional iCMs through the p38 mitogen-activated protein kinase and phosphoinositol 3-kinase/AKT pathways. Moreover, FFV enabled cardiac reprogramming with only Mef2c and Tbx5 through the induction of cardiac reprogramming factors, including Gata4. Thus, defined culture conditions promoted the quality of cardiac reprogramming, and this finding provides new insight into the mechanism of cardiac reprogramming.


Assuntos
Técnicas de Reprogramação Celular/métodos , Reprogramação Celular , Fatores de Crescimento de Fibroblastos/metabolismo , Fibroblastos/citologia , Miócitos Cardíacos/citologia , Fator A de Crescimento do Endotélio Vascular/metabolismo , Animais , Células Cultivadas , Fibroblastos/metabolismo , Fatores de Transcrição MEF2/genética , Fatores de Transcrição MEF2/metabolismo , Camundongos , Miócitos Cardíacos/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais , Proteínas com Domínio T/genética , Proteínas com Domínio T/metabolismo , Regulação para Cima , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
12.
Circ Res ; 116(8): 1378-91, 2015 Apr 10.
Artigo em Inglês | MEDLINE | ID: mdl-25858064

RESUMO

The discovery of induced pluripotent stem cells changed the field of regenerative medicine and inspired the technological development of direct reprogramming or the process by which one cell type is directly converted into another without reverting a stem cell state by overexpressing lineage-specific factors. Indeed, direct reprogramming has proven sufficient in yielding a diverse range of cell types from fibroblasts, including neurons, cardiomyocytes, endothelial cells, hematopoietic stem/progenitor cells, and hepatocytes. These studies revealed that somatic cells are more plastic than anticipated, and that transcription factors, microRNAs, epigenetic factors, secreted molecules, as well as the cellular microenvironment are all important for cell fate specification. With respect to the field of cardiology, the cardiac reprogramming presents as a novel method to regenerate damaged myocardium by directly converting endogenous cardiac fibroblasts into induced cardiomyocyte-like cells in situ. The first in vivo cardiac reprogramming reports were promising to repair infarcted hearts; however, the low induction efficiency of fully reprogrammed, functional induced cardiomyocyte-like cells has become a major challenge and hampered our understanding of the reprogramming process. Nevertheless, recent studies have identified several critical factors that may affect the efficiency and quality of cardiac induction and have provided new insights into the mechanisms of cardiac reprogramming. Here, we review the progress in direct reprogramming research and discuss the perspectives and challenges of this nascent technology in basic biology and clinical applications.


Assuntos
Reprogramação Celular , Cardiopatias/cirurgia , Células-Tronco Pluripotentes Induzidas/transplante , Miocárdio/patologia , Miócitos Cardíacos/transplante , Regeneração , Medicina Regenerativa/métodos , Animais , Linhagem da Célula , Regulação da Expressão Gênica , Cardiopatias/genética , Cardiopatias/metabolismo , Cardiopatias/patologia , Cardiopatias/fisiopatologia , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Miocárdio/metabolismo , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/patologia , Fenótipo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Resultado do Tratamento
13.
Circ Res ; 111(9): 1147-56, 2012 Oct 12.
Artigo em Inglês | MEDLINE | ID: mdl-22931955

RESUMO

RATIONALE: After myocardial infarction (MI), massive cell death in the myocardium initiates fibrosis and scar formation, leading to heart failure. We recently found that a combination of 3 cardiac transcription factors, Gata4, Mef2c, and Tbx5 (GMT), reprograms fibroblasts directly into functional cardiomyocytes in vitro. OBJECTIVE: To investigate whether viral gene transfer of GMT into infarcted hearts induces cardiomyocyte generation. METHODS AND RESULTS: Coronary artery ligation was used to generate MI in the mouse. In vitro transduction of GMT retrovirus converted cardiac fibroblasts from the infarct region into cardiomyocyte-like cells with cardiac-specific gene expression and sarcomeric structures. Injection of the green fluorescent protein (GFP) retrovirus into mouse hearts, immediately after MI, infected only proliferating noncardiomyocytes, mainly fibroblasts, in the infarct region. The GFP expression diminished after 2 weeks in immunocompetent mice but remained stable for 3 months in immunosuppressed mice, in which cardiac induction did not occur. In contrast, injection of GMT retrovirus into α-myosin heavy chain (αMHC)-GFP transgenic mouse hearts induced the expression of αMHC-GFP, a marker of cardiomyocytes, in 3% of virus-infected cells after 1 week. A pooled GMT injection into the immunosuppressed mouse hearts induced cardiac marker expression in retrovirus-infected cells within 2 weeks, although few cells showed striated muscle structures. To transduce GMT efficiently in vivo, we generated a polycistronic retrovirus expressing GMT separated by 2A "self-cleaving" peptides (3F2A). The 3F2A-induced cardiomyocyte-like cells in fibrotic tissue expressed sarcomeric α-actinin and cardiac troponin T and had clear cross striations. Quantitative RT-PCR also demonstrated that FACS-sorted 3F2A-transduced cells expressed cardiac-specific genes. CONCLUSIONS: GMT gene transfer induced cardiomyocyte-like cells in infarcted hearts.


Assuntos
Diferenciação Celular/genética , Fator de Transcrição GATA4/genética , Técnicas de Transferência de Genes , Infarto do Miocárdio/patologia , Miócitos Cardíacos/patologia , Fatores de Regulação Miogênica/genética , Proteínas com Domínio T/genética , Animais , Diferenciação Celular/fisiologia , Fibroblastos/patologia , Fator de Transcrição GATA4/fisiologia , Proteínas de Fluorescência Verde/genética , Fatores de Transcrição MEF2 , Masculino , Camundongos , Camundongos Endogâmicos ICR , Camundongos Nus , Camundongos Transgênicos , Modelos Animais , Infarto do Miocárdio/fisiopatologia , Miócitos Cardíacos/fisiologia , Fatores de Regulação Miogênica/fisiologia , Regeneração/genética , Regeneração/fisiologia , Retroviridae/genética , Proteínas com Domínio T/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA