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










Base de dados
Intervalo de ano de publicação
3.
Cell Prolif ; 50(1)2017 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-27790820

RESUMO

OBJECTIVES: Induced pluripotent stem cell (iPSC)-derived cardiomyocytes are a promising source of cells for regenerative heart disease therapies, but progress towards their use has been limited by their low differentiation efficiency and high cellular heterogeneity. Previous studies have demonstrated expression of adrenergic receptors (ARs) in stem cells after differentiation; however, roles of ARs in fate specification of stem cells, particularly in cardiomyocyte differentiation and development, have not been characterized. MATERIALS AND METHODS: Murine-induced pluripotent stem cells (miPSCs) were cultured in hanging drops to form embryoid bodies, cells of which were then differentiated into cardiomyocytes. To determine whether ARs regulated miPSC differentiation into cardiac lineages, effects of the AR agonist, epinephrine (EPI), on miPSC differentiation and underlying signalling mechanisms, were evaluated. RESULTS: Treatment with EPI, robustly enhanced miPSC cardiac differentiation, as indicated by increased expression levels of cardiac-specific markers, GATA4, Nkx2.5 and Tnnt2. Although ß-AR signalling is the foremost signalling pathway in cardiomyocytes, EPI-enhanced cardiac differentiation depended more on α-AR signalling than ß-AR signalling. In addition, selective activation of α1 -AR signalling with specific agonists induced vigorous cardiomyocyte differentiation, whereas selective activation of α2 - or ß-AR signalling induced no or less differentiation, respectively. EPI- and α1 -AR-dependent cardiomyocyte differentiation from miPSCs occurred through specific promotion of CPC proliferation via the MEK-ERK1/2 pathway and regulation of miPS cell-cycle progression. CONCLUSIONS: These results demonstrate that activation of ARs, particularly of α1 -ARs, promoted miPSC differentiation into cardiac lineages via MEK-ERK1/2 signalling.


Assuntos
Diferenciação Celular , Células-Tronco Pluripotentes Induzidas/citologia , Miócitos Cardíacos/citologia , Receptores Adrenérgicos alfa/metabolismo , Receptores Adrenérgicos beta/metabolismo , Agonistas Adrenérgicos beta/farmacologia , Antagonistas Adrenérgicos beta/farmacologia , Animais , Apoptose , Pontos de Checagem do Ciclo Celular , Diferenciação Celular/efeitos dos fármacos , Proliferação de Células , Células Cultivadas , Corpos Embrioides/metabolismo , Epinefrina/farmacologia , Fator de Transcrição GATA4/metabolismo , Proteína Homeobox Nkx-2.5/metabolismo , Células-Tronco Pluripotentes Induzidas/efeitos dos fármacos , Células-Tronco Pluripotentes Induzidas/metabolismo , Camundongos , Microscopia de Fluorescência , Miócitos Cardíacos/metabolismo , Receptores Adrenérgicos beta/química , Transdução de Sinais/efeitos dos fármacos , Troponina T/metabolismo
4.
Exp Cell Res ; 345(1): 25-36, 2016 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-27208585

RESUMO

Diabetic cardiomyopathy increases the risk for the development of heart failure independent of coronary artery disease and hypertension. Either type 1 or type 2 diabetes is often accompanied by varying degrees of hyperglycemia, which has been proven to induce myocardial apoptosis in animal models. Recently, a novel small molecule, ZLN005, has been reported to show antidiabetic efficacy in a mouse model, possibly by induction of PGC-1α expression. In this study, we investigated whether ZLN005 protects cardiomyocytes against high glucose-induced cytotoxicity and the mechanisms involved. Neonatal mouse cardiomyocytes were incubated with media containing 5.5 or 33mM glucose for 24h in the presence or absence of ZLN005. ZLN005 treatment led to ameliorated cardiomyocyte oxidative injury, enhanced cell viability, and reduced apoptosis in the high glucose environment. Western blot analysis revealed that high glucose suppressed cardiomyocyte autophagy, whereas ZLN005 increased the expression of autophagy marker proteins ATG5, beclin1, and LC3 II/LC3 I; this increase was accompanied by increased expression of SIRT1. Furthermore, EX527, a SIRT1-specific inhibitor, weakened the protective effects of ZLN005 on cardiomyocytes subjected to high glucose. Taken together, these results suggest that ZLN005 suppresses high glucose-induced cardiomyocyte injury by promoting SIRT1 expression and autophagy.


Assuntos
Autofagia/efeitos dos fármacos , Benzimidazóis/farmacologia , Cardiotônicos/farmacologia , Glucose/toxicidade , Miócitos Cardíacos/citologia , Sirtuína 1/genética , Animais , Animais Recém-Nascidos , Apoptose/efeitos dos fármacos , Autofagia/genética , Sobrevivência Celular/efeitos dos fármacos , Citoproteção/efeitos dos fármacos , Estresse do Retículo Endoplasmático/efeitos dos fármacos , L-Lactato Desidrogenase/metabolismo , Camundongos Endogâmicos C57BL , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/metabolismo , Estresse Oxidativo/efeitos dos fármacos , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Sirtuína 1/metabolismo
5.
BMC Dev Biol ; 16: 11, 2016 May 04.
Artigo em Inglês | MEDLINE | ID: mdl-27141946

RESUMO

BACKGROUND: We previously reported that the pluripotent stem cells can differentiate into cardiomyocytes (CMs) by co-culture with neonatal CMs (NCMs) in vitro. However, the involving mechanism is not clear. METHODS: Mouse induced pluripotent stem cells (iPSCs) were cultured in hanging drops to form embryoid bodies (EBs) and to induce myocardial differentiation. Co-culture of EBs and NCMs was established in a transwell insert system, while EBs grown alone in the wells were used as controls. RESULTS: Co-culture with NCMs markedly increased the generation of functional CMs from iPSCs. The focal adhesion kinase (FAK) phosphorylation, and c-Jun N-terminal kinase (JNK) phosphorylation in co-culture were higher than that in EBs grown alone. Treating FAK small interfering RNA (FAK siRNA) or specific inhibitor for JNK (SP600125) to iPSCs significantly reduced the phosphorylation of JNK and the expressions of Mef2c and Bcl-2. The expressions of cTnT and MLC-2V were also decreased. Our results revealed that co-culture with NCMs significantly enhance the differentiation ability of iPSCs by increasing Mef2c and Bcl-2 expressions concomitantly with a marked augment on cell proliferation through JNK signaling pathways. CONCLUSIONS: These findings indicated that co-culture of EBs with NCMs induces genes expressed in a mature pattern and stimulates the proliferation of iPSC-derived CMs (iPS-CMs) by activating FAK/JNK signaling.


Assuntos
Técnicas de Cocultura/métodos , Proteína-Tirosina Quinases de Adesão Focal/metabolismo , Células-Tronco Pluripotentes Induzidas/citologia , Sistema de Sinalização das MAP Quinases , Miócitos Cardíacos/citologia , Miócitos Cardíacos/enzimologia , Animais , Animais Recém-Nascidos , Diferenciação Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Células-Tronco Pluripotentes Induzidas/efeitos dos fármacos , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Camundongos , Miocárdio/citologia , Miócitos Cardíacos/efeitos dos fármacos , Especificidade de Órgãos/efeitos dos fármacos , Inibidores de Proteínas Quinases/farmacologia , RNA Interferente Pequeno/metabolismo
6.
PLoS One ; 8(1): e55233, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23383121

RESUMO

BACKGROUND: Embryonic Stem Cells (ESCs) can differentiate into cardiomyocytes (CMs) in vitro but the differentiation level from ESCs is low. Here we describe a simple co-culture model by commercially available Millicell™ hanging cell culture inserts to control the long-term differentiation of ESCs into CMs. METHODOLOGY/PRINCIPAL FINDINGS: Mouse ESCs were cultured in hanging drops to form embryoid bodies (EBs) and treated with 0.1 mmol/L ascorbic acid to induce the differentiation of ESCs into CMs. In the indirect co-culture system, EBs were co-cultured with epidermal keratinocytes (EKs) or neonatal CMs (NCMs) by the hanging cell culture inserts (PET membranes with 1 µm pores). The molecular expressions and functional properties of ESC-derived CMs in prolonged culture course were evaluated. During time course of ESC differentiation, the percentages of EBs with contracting areas in NCMs co-culture were significantly higher than that without co-culture or in EKs co-culture. The functional maintenance of ESC-derived CMs were more prominent in NCMs co-culture model. CONCLUSIONS/SIGNIFICANCE: These results indicate that NCMs co-culture promote ESC differentiation and has a further effect on cell growth and differentiation. We assume that the improvement of the differentiating efficiency of ESCs into CMs in the co-culture system do not result from the effect of co-culture directly on cell differentiation, but rather by signaling effects that influence the cells in proliferation and long-term function maintenance.


Assuntos
Técnicas de Cultura de Células/métodos , Diferenciação Celular/fisiologia , Células-Tronco Embrionárias/citologia , Miócitos Cardíacos/citologia , Animais , Ácido Ascórbico/farmacologia , Diferenciação Celular/efeitos dos fármacos , Técnicas de Cocultura/métodos , Camundongos
7.
BMC Cell Biol ; 14: 5, 2013 Jan 25.
Artigo em Inglês | MEDLINE | ID: mdl-23350814

RESUMO

BACKGROUND: The interactions between stem cells and extracellular matrix (ECM) mediated by integrins play important roles in the processes that determine stem cell fate. However, the role of ECM/integrin interaction in the formation of embryoid bodies (EBs) during cardiogenesis from murine induced pluripotent stem cells (miPSCs) remains unclear. RESULTS: In the present study, collagen type I and ß(1) integrin were expressed and upregulated synergistically during the formation of miPSC-derived EBs, with a peak expression at day 3 of differentiation. The blockage of collagen/ß(1) integrin interaction by ß(1) integrin blocking antibody resulted in the production of defective EBs that were characterized by decreased size and the absence of a shell-like layer composed of primitive endoderm cells. The quantification of spontaneous beating activity, cardiac-specific gene expression and cardiac troponin T (cTnT) immunostaining showed that the cardiac differentiation of these defective miPSC-derived EBs was lower than that of control EBs. CONCLUSIONS: These findings indicate that collagen/ß(1) integrin interaction is required for the growth and cardiac differentiation of miPSC-derived EBs and will be helpful in future engineering of the matrix microenvironment within EBs to efficiently direct the cardiac fate of pluripotent stem cells to promote cardiovascular regeneration.


Assuntos
Colágeno Tipo I/metabolismo , Corpos Embrioides/citologia , Células-Tronco Pluripotentes Induzidas/metabolismo , Integrina beta1/metabolismo , Animais , Anticorpos/imunologia , Diferenciação Celular , Células Cultivadas , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/metabolismo , Imuno-Histoquímica , Células-Tronco Pluripotentes Induzidas/citologia , Integrina beta1/imunologia , Camundongos , Microscopia Eletrônica de Varredura , Miocárdio/citologia , Ligação Proteica , Troponina T/metabolismo
8.
Int J Biol Sci ; 8(2): 236-48, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22253567

RESUMO

OBJECTIVES: Hypertrophy has been shown to be associated with arrhythmias which can be caused by abnormal remodeling of the Kv4-family of transient potassium channels. Inhibitors of 3-hydroxy-3-methylglutaryl coenzyme A reductase (statins) have recently been shown to exert pleiotropic protective effects in cardiovascular diseases, including anti-arrhythmias. It is hypothesized that remodeling of Kv4.3 occurs in rat hypertrophied cardiomyocytes and is regulated by simvastatin. METHODS: Male Sprague-Dawley rats and neonatal rat ventricular myocytes (NRVMs) underwent abdominal aortic banding (AAB) for 7 weeks and angiotensin II (AngII) treatment, respectively, to induce cardiac hypertrophy. Kv4.3 expression by NRVMs and myocardium (subepicardial and subendocardial) in the left ventricle was measured. The transient outward potassium current (I(to)) of NRVMs was recorded using a whole-cell patch-clamp method. RESULTS: Expression of the Kv4.3 transcript and protein was significantly reduced in myocardium (subepicardial and subendocardial) in the left ventricle and in NRVMs. Simvastatin partially prevented the reduction of Kv4.3 expression in NRVMs and subepicardial myocardium but not in the subendocardial myocardium. Hypertrophied NRVMs exhibited a significant reduction in the I(to) current and this effect was partially reversed by simvastatin. CONCLUSIONS: Simvastatin alleviated the reduction of Kv4.3 expression, I(to) currents in hypertrophied NRVMs and alleviated the reduced Kv4.3 expression in subepicardial myocardium from the hypertrophied left ventricle. It can be speculated that among the pleiotropic effects of simvastatin, the anti-arrhythmia effect is partly mediated by its effect on Kv4.3.


Assuntos
Regulação da Expressão Gênica/efeitos dos fármacos , Inibidores de Hidroximetilglutaril-CoA Redutases/farmacologia , Miócitos Cardíacos/efeitos dos fármacos , Canais de Potássio Shal/metabolismo , Sinvastatina/farmacologia , Animais , Calcineurina/metabolismo , Cardiomegalia/tratamento farmacológico , Cardiomegalia/patologia , Metabolismo dos Lipídeos/efeitos dos fármacos , Masculino , Miócitos Cardíacos/metabolismo , Ratos , Ratos Sprague-Dawley , Canais de Potássio Shal/genética
9.
J Cell Biochem ; 112(12): 3555-62, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21780160

RESUMO

The cardiomyocyte (CM) differentiation of embryonic stem cells (ESCs) is routinely cultured as two-dimensional (2D) monolayer, which doesn't mimic in vivo physiological environment and may lead to low differentiated level of ESCs. Here, we develop a novel strategy that enhances CM differentiation of ESCs in collagen matrix three-dimensional (3D) culture combined with indirect cardiac fibroblasts co-culture. ESCs were cultured in hanging drops to form embryoid bodies (EBs) and then applied on collagen matrix. The EBs were indirectly co-cultured with cardiac fibroblasts by the hanging cell culture inserts (PET 1 µm). The molecular expressions and ultrastructural characteristics of ESC-derived CMs (ESCMs) were analyzed by real time RT-PCR, immunocytochemistry, and Transmission Electron Microscopy (TEM). We found that the percentage of beating EBs with cardiac fibroblasts co-culture was significantly higher than that without co-culture after differentiation period of 8 days. Type I collagen used as 3D substrates enhanced the late-stage CM differentiation of ESCs and had effect on ultrastructural mature of ESCMs in late-stage development. The combined effects of 3D and co-culture that mimic in vivo physiological environment further improved the efficiency of CM differentiation from ESCs, resulting in fiber-like structures of cardiac cells with organized sarcomeric structure in ESCMs. This novel 3D co-culture system emphasizes the fact that the ESC differentiation is actively responding to cues from their environment and those cues can drive phenotypic control, which provides a useful in vitro model to investigate CM differentiation of stem cells.


Assuntos
Diferenciação Celular , Células-Tronco Embrionárias/citologia , Miocárdio/citologia , Animais , Sequência de Bases , Técnicas de Cocultura , Primers do DNA , Células-Tronco Embrionárias/ultraestrutura , Imuno-Histoquímica , Camundongos , Microscopia Eletrônica de Transmissão , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa
13.
Cell Biol Int ; 2009 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-19947942

RESUMO

We studied the differentiation of embryonic stem cells (ESCs) and developed a novel protocol for generating functional cardiomyocytes (CMs) from ESCs by co-culturing these with live cardiac cells. We then evaluated the structural and functional properties of these ESC-derived CMs (ESCMs). An acellular matrix obtained from rabbit heart tissues was used as a scaffold. Then ESCMs were seeded onto the acellular matrix for preliminary tissue engineering applications. We found that by mimicking the cardiac microenvironment, the percentage of beating embryoid bodies (EBs) was much higher and the homogeneity of EBs were significantly improved over that seen in the control group (p<0.001). ESCMs in EBs acquired almost the same structural and functional properties as typical CMs. After implantation, the cells in the EBs rapidly grew and expanded in the extracellular matrix. These results indicate that the differentiation of ESCs can be controlled in a cardiac mimicking microenvironment and that ESCs can be used as an ideal cell source for large-scale tissue engineering applications for the procurement of cardiac muscle.

14.
Cell Biol Int ; 33(10): 1123-4, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19545639

RESUMO

This article gives a brief comment on the culture of human embryonic stem cells (hESCs) with the aim to maintain the potency of hESCs in well state and produce more homogenous cell clones.


Assuntos
Técnicas de Cultura de Células , Células-Tronco Embrionárias/fisiologia , Células Clonais , Humanos
15.
Bioessays ; 31(2): 246-52, 2009 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19204979

RESUMO

Biological pacemakers can be achieved by various gene-based and cell-based approaches. Embryonic stem cells (ESCs)-derived pacemaker cells might be the most promising way to form biological pacemakers, but there are challenges as to how to control the differentiation of ESCs and to overcome the neoplasia, proarrhythmia, or immunogenicity resulting from the use of ESCs. As a potential approach to solve these difficult problems, tissue-engineering techniques may provide a precise control on the different cell components of multicellular aggregates and the forming of a construct with-defined architectures and functional properties. The combined interactions between ESC-derived pacemaker cells, supporting cells, and matrices may completely reproduce pacemaker properties and result in a steady functional unit to induce rhythmic electrical and contractile activities. As ESCs have a high capability for self-renewal, proliferation, and potential differentiation, we hypothesize that ESCs can be used as a source of pacemaker cells for tissue-engineering applications and the ambitious goal of biological cardiac pacemakers may ultimately be achieved with ESCs via tissue-engineering technology.


Assuntos
Relógios Biológicos , Células-Tronco Embrionárias/metabolismo , Engenharia Tecidual , Animais , Humanos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA