Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 77
Filtrar
1.
Int J Mol Sci ; 22(17)2021 Aug 25.
Artigo em Inglês | MEDLINE | ID: mdl-34502068

RESUMO

Although advances in rapid revascularization strategies following acute myocardial infarction (AMI) have led to improved short and long-term outcomes, the associated loss of cardiomyocytes and the subsequent remodeling result in an impaired ventricular function that can lead to heart failure or death. The poor regenerative capacity of the myocardium and the current lack of effective regenerative therapies have driven stem cell research in search of a possible solution. One approach involves the delivery of stem cells to the site of injury in order to stimulate repair response. Although animal studies initially delivered promising results, the application of similar techniques in humans has been hampered by poor target site retention and oncogenic considerations. In response, several alternative strategies, including the use of non-coding RNAs (ncRNAs), have been introduced with the aim of activating and regulating stem cells or inducing stem cell status in resident cells. Circular RNAs (circRNAs) and microRNAs (miRNAs) are ncRNAs with pivotal functions in cell proliferation and differentiation, whose role in stem cell regulation and potential significance for the field of cardiac regeneration is the primary focus of this review. We also address the general advantages of ncRNAs as promising drivers of cardiac regeneration and potent stem cell regulators.


Assuntos
MicroRNAs/metabolismo , Mioblastos Cardíacos/metabolismo , RNA Longo não Codificante/metabolismo , Regeneração , Animais , Diferenciação Celular , Humanos , MicroRNAs/genética , Mioblastos Cardíacos/citologia , Mioblastos Cardíacos/fisiologia , RNA Longo não Codificante/genética
2.
Metabolism ; 121: 154778, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-33901502

RESUMO

Glutamine is a major energy source for rapidly dividing cells, such as hematopoietic stem cells and cancer cells. Reliance on glutamine is therefore regarded as a metabolic hallmark of proliferating cells. Moreover, reprogramming glutamine metabolism by various factors, including tissue type, microenvironment, pro-oncogenes, and tumor suppressor genes, can facilitate stem cell fate decisions, tumor recurrence, and drug resistance. However, the significance of glutamine metabolism in cardiomyocytes, an end-differentiated cell type, is not fully understood. Existing evidence suggests important roles of glutamine metabolism in the development of cardiovascular diseases. In this review, we have focused on glutaminolysis and its regulatory network in proliferating cells. We have summarized current findings about the role of glutamine utilization in cardiomyocytes and have discussed possibilities of targeting glutamine metabolism for the treatment of cardiovascular diseases.


Assuntos
Proliferação de Células/fisiologia , Glutamina/metabolismo , Miócitos Cardíacos/metabolismo , Animais , Diferenciação Celular/fisiologia , Humanos , Mioblastos Cardíacos/fisiologia , Neoplasias/metabolismo , Neoplasias/patologia , Células-Tronco/fisiologia , Microambiente Tumoral
3.
Sci Rep ; 10(1): 1895, 2020 02 05.
Artigo em Inglês | MEDLINE | ID: mdl-32024875

RESUMO

The possibility of using stem cell-derived cardiomyocytes opens a new platform for modeling cardiac cell differentiation and disease or the development of new drugs. Progress in this field can be accelerated by high-throughput screening (HTS) technology combined with promoter reporter system. The goal of the study was to create and evaluate a responsive promoter reporter system that allows monitoring of iPSC differentiation towards cardiomyocytes. The lentiviral promoter reporter system was based on troponin 2 (TNNT2) and alpha cardiac actin (ACTC) with firefly luciferase and mCherry, respectively. The system was evaluated in two in vitro models. First, system followed the differentiation of TNNT2-luc-T2A-Puro-mCMV-GFP and hACTC-mcherry-WPRE-EF1-Neo from transduced iPSC line towards cardiomyocytes and revealed the significant decrease in both inserts copy number during the prolonged in vitro cell culture (confirmed by I-FISH, ddPCR, qPCR). Second, differentiated and contracting control cardiomyocytes (obtained from control non-reporter transduced iPSCs) were subsequently transduced with TNNT2-luc-T2A-Puro-CMV-GFP and hACTC-mcherry-WPRE-EF1-Neo lentiviruses to observe the functionality of obtained cardiomyocytes. Our results indicated that the reporter modified cell lines can be used for HTS applications, but it is essential to monitor the stability of the reporter sequence during extended cell in vitro culture.


Assuntos
Diferenciação Celular , Ensaios de Triagem em Larga Escala/métodos , Células-Tronco Pluripotentes Induzidas/fisiologia , Mioblastos Cardíacos/fisiologia , Miócitos Cardíacos/fisiologia , Actinas/genética , Adulto , Células Cultivadas , Técnicas de Reprogramação Celular/métodos , Genes Reporter/genética , Humanos , Lentivirus/genética , Luciferases de Vaga-Lume/genética , Proteínas Luminescentes/genética , Masculino , Cultura Primária de Células , Regiões Promotoras Genéticas/genética , Transdução Genética , Troponina T/genética , Proteína Vermelha Fluorescente
4.
BMC Mol Cell Biol ; 20(1): 25, 2019 07 17.
Artigo em Inglês | MEDLINE | ID: mdl-31315556

RESUMO

BACKGROUND: Cardiac stem cells (CSCs) exhibit age-dependent characteristics. Cited2 has been implicated in the regulation of heart development; however, there is little known about how Cited2 affects CSC aging. RESULTS: Cited2 mRNA and protein level was downregulated in aging heart tissue and CSCs. Old (O)-CSCs showed decreased differentiation and proliferation capacities as compared to Young (Y)-CSCs, the decrease in cell proliferation, increase in apoptosis, and cell cycle arrest in G0/G1 phase in CSCs are mediated by knocdown CITED2expression in (Y)-CSCs. CONCLUSIONS: Cited2 plays an important role in cell cycle progression and in maintaining the balance between CSC proliferation and apoptosis in the process of aging without influencing cell fate decisions. These findings have important implications for cell-based therapies for heart repair.


Assuntos
Proliferação de Células/fisiologia , Sobrevivência Celular/fisiologia , Senescência Celular/fisiologia , Mioblastos Cardíacos/fisiologia , Proteínas Repressoras/genética , Proteínas Repressoras/metabolismo , Transativadores/genética , Transativadores/metabolismo , Animais , Apoptose/fisiologia , Diferenciação Celular , Terapia Baseada em Transplante de Células e Tecidos , Pontos de Checagem da Fase G1 do Ciclo Celular , Regulação da Expressão Gênica no Desenvolvimento , Técnicas de Silenciamento de Genes , Camundongos , Camundongos Endogâmicos C57BL , Mioblastos Cardíacos/citologia , RNA Interferente Pequeno/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transcriptoma , Transfecção
6.
Apoptosis ; 24(3-4): 359-368, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30737648

RESUMO

During the development of cardiac hypertrophy, glucose deprivation (GD) associated with coronary microvascular rarefaction is caused, leading to cardiomyocyte death. Phosphorylation (inactivation) of eukaryotic elongation factor 2 (eEF2) by eEF2 kinase (eEF2K) inhibits protein translation, a highly energy consuming process, which plays protective roles against nutrient deprivation-induced cell death. We previously showed that eEF2 phosphorylation was increased in isolated heart from several cardiac hypertrophy models. In this study, we investigated whether eEF2K/eEF2 mediates the inhibition of cardiomyocyte death under GD condition. In H9c2 rat cardiomyoblasts cultured with serum-free medium, GD significantly augmented eEF2 phosphorylation and signals related to autophagy [increase of microtubule-associated protein 1 light chain 3 (LC3)-II to LC3-I ratio] and apoptosis (cleavage of caspase-3) as determined by Western blotting. GD induced cell death, which was augmented by eEF2K gene knockdown using a small interfering RNA. eEF2K gene knockdown significantly augmented GD-induced cleavage of caspase-3 and apoptotic nuclear condensation as determined by 4', 6-diamidino-2-phenylindole staining. In contrast, eEF2K gene knockdown significantly inhibited GD-induced increase of LC3-II to LC3-I ratio and autophagosome formation as determined by an immunofluorescence staining. An inhibitor of autophagy, 3-methyladenine or bafilomycin A1 significantly augmented GD-induced cleavage of caspase-3. Further, eEF2K gene knockdown significantly inhibited GD-induced phosphorylation of adenosine monophosphate-activated protein kinase (AMPK)α and its downstream substrate, unc-51 like autophagy activating kinase (ULK)1. An inhibitor of AMPK, dorsomorphin significantly inhibited GD-induced increase of LC3-II to LC3-I ratio. In conclusion, we for the first time revealed that eEF2K/eEF2 axis under GD condition mediates the inhibition of apoptotic H9c2 cell death at least in part via promotion of autophagy through AMPKα/ULK1 signaling pathway.


Assuntos
Morte Celular/fisiologia , Quinase do Fator 2 de Elongação/metabolismo , Glucose/metabolismo , Mioblastos Cardíacos/metabolismo , Fator 2 de Elongação de Peptídeos/metabolismo , Proteínas Quinases Ativadas por AMP/metabolismo , Animais , Apoptose/fisiologia , Autofagossomos/metabolismo , Autofagia/fisiologia , Caspase 3/metabolismo , Linhagem Celular , Mioblastos Cardíacos/fisiologia , Fosforilação/fisiologia , Ratos , Transdução de Sinais/fisiologia
7.
Math Med Biol ; 35(1): 121-144, 2018 03 14.
Artigo em Inglês | MEDLINE | ID: mdl-28115549

RESUMO

We propose a discrete in continuous mathematical model describing the in vitro growth process of biophsy-derived mammalian cardiac progenitor cells growing as clusters in the form of spheres (Cardiospheres). The approach is hybrid: discrete at cellular scale and continuous at molecular level. In the present model, cells are subject to the self-organizing collective dynamics mechanism and, additionally, they can proliferate and differentiate, also depending on stochastic processes. The two latter processes are triggered and regulated by chemical signals present in the environment. Numerical simulations show the structure and the development of the clustered progenitors and are in a good agreement with the results obtained from in vitro experiments.


Assuntos
Fenômenos Fisiológicos Celulares/fisiologia , Modelos Teóricos , Mioblastos Cardíacos/fisiologia , Esferoides Celulares/fisiologia , Animais , Humanos
9.
Sci Rep ; 6: 30922, 2016 08 04.
Artigo em Inglês | MEDLINE | ID: mdl-27488808

RESUMO

Research has demonstrated that hypoxic preconditioning (HP) can enhance the survival and proliferation of cardiac progenitor cells (CPCs); however, the underlying mechanisms are not fully understood. Here, we report that HP of c-kit (+) CPCs inhibits p53 via the PI3K/Akt-DNMT1 pathway. First, CPCs were isolated from the hearts of C57BL/6 mice and further purified by magnetic-activated cell sorting. Next, these cells were cultured under either normoxia (H0) or HP for 6 hours (H6) followed by oxygen-serum deprivation for 24 hours (24h). Flow cytometric analysis and MTT assays revealed that hypoxia-preconditioned CPCs exhibited an increased survival rate. Western blot and quantitative real-time PCR assays showed that p53 was obviously inhibited, while DNMT1 and DNMT3ß were both significantly up-regulated by HP. Bisulphite sequencing analysis indicated that DNMT1 and DNMT3ß did not cause p53 promoter hypermethylation. A reporter gene assay and chromatin immunoprecipitation analysis further demonstrated that DNMT1 bound to the promoter locus of p53 in hypoxia-preconditioned CPCs. Together, these observations suggest that HP of CPCs could lead to p53 inhibition by up-regulating DNMT1 and DNMT3ß, which does not result in p53 promoter hypermethylation, and that DNMT1 might directly repress p53, at least in part, by binding to the p53 promoter locus.


Assuntos
Hipóxia/terapia , Precondicionamento Isquêmico Miocárdico , Mioblastos Cardíacos/fisiologia , Animais , Apoptose , Proliferação de Células , Células Cultivadas , DNA (Citosina-5-)-Metiltransferase 1/metabolismo , Metilação de DNA , Hipóxia/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Proteína Oncogênica v-akt/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Regiões Promotoras Genéticas/genética , Proteína Supressora de Tumor p53/metabolismo
10.
Sci Rep ; 6: 30956, 2016 08 05.
Artigo em Inglês | MEDLINE | ID: mdl-27492371

RESUMO

Efficient generation of cardiomyocytes from human pluripotent stem cells is critical for their regenerative applications. Microgravity and 3D culture can profoundly modulate cell proliferation and survival. Here, we engineered microscale progenitor cardiac spheres from human pluripotent stem cells and exposed the spheres to simulated microgravity using a random positioning machine for 3 days during their differentiation to cardiomyocytes. This process resulted in the production of highly enriched cardiomyocytes (99% purity) with high viability (90%) and expected functional properties, with a 1.5 to 4-fold higher yield of cardiomyocytes from each undifferentiated stem cell as compared with 3D-standard gravity culture. Increased induction, proliferation and viability of cardiac progenitors as well as up-regulation of genes associated with proliferation and survival at the early stage of differentiation were observed in the 3D culture under simulated microgravity. Therefore, a combination of 3D culture and simulated microgravity can be used to efficiently generate highly enriched cardiomyocytes.


Assuntos
Simulação por Computador , Mioblastos Cardíacos/fisiologia , Miócitos Cardíacos/fisiologia , Células-Tronco Pluripotentes/fisiologia , Diferenciação Celular , Proliferação de Células , Sobrevivência Celular , Células Cultivadas , Humanos , Técnicas de Cultura de Órgãos , Engenharia Tecidual , Simulação de Ausência de Peso
11.
Int J Cardiol ; 221: 396-403, 2016 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-27404713

RESUMO

In the heart, tissue-derived signals play a central role on recruiting/activating stem cell sources to induce cardiac lineage specification for maintenance of tissue homeostasis and repair. Cardiac resident stromal cells (CRSCs) may play a pivotal role in cardiac repair throughout their secretome. Here, we performed the characterization of CRSCs and their secretome by analyzing the composition of their culture-derived extracellular matrix (ECM) and conditioned medium (CM) and by investigating their potential effect on adipose-derived stem cell (ADSC) and progenitor cell behavior. We confirmed that CRSCs are a heterogeneous cell population whose secretome is composed by proteins related to cellular growth, immune response and cardiovascular development and function. We also observed that CRSC secretome was unable to change the behavior of ADSCs, except for proliferation. Additionally, CM from CRSCs demonstrated the potential to drive proliferation and cardiac differentiation of H9c2 cells and also the ability to induce angiogenesis in vitro. Our data suggest that the CRSCs can be a source of important modulating signals for cardiac progenitor cell recruitment/activation.


Assuntos
Tecido Adiposo/citologia , Mioblastos Cardíacos/fisiologia , Miocárdio/citologia , Via Secretória/fisiologia , Diferenciação Celular , Proliferação de Células , Células Cultivadas , Meios de Cultivo Condicionados , Matriz Extracelular , Humanos , Células Estromais/fisiologia
12.
J Surg Res ; 203(2): 483-90, 2016 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-27363659

RESUMO

BACKGROUND: Hyperglycemia is proposed to be an independent risk factor for cardiovascular morbidity and mortality. Preclinical studies suggest that diabetes mellitus exacerbates myocardial ischemia/reperfusion injury and attenuates the effects of cardioprotective strategies. The cardioprotective effects of postconditioning with the opioid analgesic remifentanil against ischemia/reperfusion injury under the hyperglycemic condition remain contradictory. Therefore, the aim of this study was to investigate the mechanisms by which hyperglycemia affects cardioprotection induced by remifentanil postconditioning. MATERIALS AND METHODS: H9c2 cardiomyoblasts were cultured under the normoglycemic or hyperglycemic condition. Cells were exposed to hypoxia/reoxygenation (H/R) injury followed by hypoxia postconditioning (HPC group) or remifentanil postconditioning (RPC group). Cell viability, injury, and apoptosis were measured after each postconditioning treatment. Activation of endoplasmic reticulum stress (ERS) was analyzed by examining the protein levels of GRP78, CHOP, cleaved caspase-12 and cleaved caspase-3. RESULTS: RPC significantly increased cell viability and reduced apoptosis in normoglycemic cardiomyoblasts, but not in hyperglycemic cardiomyoblasts. HPC and RPC markedly decreased the upregulation of GRP78, CHOP, cleaved caspase 12, and cleaved caspase 3 in response to H/R injury under the normoglycemic condition. Hyperglycemia significantly increased these ERS-associated biomarkers and apoptosis, which could not be reduced by HPC or RPC. CONCLUSIONS: Remifentanil postconditioning protected cardiomyoblasts from H/R injury under normoglycemia, at least in part, through inhibiting ERS-induced apoptosis. Hyperglycemia attenuated the cardioprotection conferred by remifentanil postconditioning, likely as a result of the exacerbated ERS. Inhibiting the ERS response may be an attractive strategy to enhance the cardioprotective effects of postconditioning in diabetic patients.


Assuntos
Analgésicos Opioides/uso terapêutico , Hiperglicemia/complicações , Pós-Condicionamento Isquêmico/métodos , Mioblastos Cardíacos/fisiologia , Traumatismo por Reperfusão Miocárdica/terapia , Piperidinas/uso terapêutico , Analgésicos Opioides/farmacologia , Apoptose/efeitos dos fármacos , Biomarcadores/metabolismo , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Chaperona BiP do Retículo Endoplasmático , Humanos , Hiperglicemia/metabolismo , Mioblastos Cardíacos/efeitos dos fármacos , Traumatismo por Reperfusão Miocárdica/complicações , Traumatismo por Reperfusão Miocárdica/metabolismo , Piperidinas/farmacologia , Remifentanil
13.
Int J Mol Sci ; 16(10): 25199-213, 2015 Oct 23.
Artigo em Inglês | MEDLINE | ID: mdl-26512644

RESUMO

Cardiomyocyte progenitor cells play essential roles in early heart development, which requires highly controlled cellular organization. microRNAs (miRs) are involved in various cell behaviors by post-transcriptional regulation of target genes. However, the roles of miRNAs in human cardiomyocyte progenitor cells (hCMPCs) remain to be elucidated. Our previous study showed that miR-134 was significantly downregulated in heart tissue suffering from congenital heart disease, underlying the potential role of miR-134 in cardiogenesis. In the present work, we showed that the upregulation of miR-134 reduced the proliferation of hCMPCs, as determined by EdU assay and Ki-67 immunostaining, while the inhibition of miR-134 exhibited an opposite effect. Both up- and downregulation of miR-134 expression altered the transcriptional level of cell-cycle genes. We identified Meis2 as the target of miR-134 in the regulation of hCMPC proliferation through bioinformatic prediction, luciferase reporter assay and western blot. The over-expression of Meis2 mitigated the effect of miR-134 on hCMPC proliferation. Moreover, miR-134 did not change the degree of hCMPC differentiation into cardiomyocytes in our model, suggesting that miR-134 is not required in this process. These findings reveal an essential role for miR-134 in cardiomyocyte progenitor cell biology and provide new insights into the physiology and pathology of cardiogenesis.


Assuntos
Proliferação de Células , Proteínas de Homeodomínio/metabolismo , MicroRNAs/genética , Mioblastos Cardíacos/metabolismo , Miócitos Cardíacos/metabolismo , Fatores de Transcrição/metabolismo , Células Cultivadas , Proteínas de Homeodomínio/genética , Humanos , Mioblastos Cardíacos/fisiologia , Miócitos Cardíacos/fisiologia , Fatores de Transcrição/genética
14.
Compr Physiol ; 5(4): 1775-814, 2015 Sep 20.
Artigo em Inglês | MEDLINE | ID: mdl-26426466

RESUMO

Cardiac aging has been confounded by the concept that the heart is a postmitotic organ characterized by a predetermined number of myocytes, which is established at birth and largely preserved throughout life until death of the organ and organism. Based on this premise, the age of cardiac cells should coincide with that of the organism; at any given time, the heart would be composed of a homogeneous population of myocytes of identical age. The discovery that stem cells reside in the heart and generate cardiac cell lineages has imposed a reconsideration of the mechanisms implicated in the manifestations of the aging myopathy. The progressive alterations of terminally differentiated myocytes, and vascular smooth muscle cells and endothelial cells may represent an epiphenomenon dictated by aging effects on cardiac progenitor cells (CPCs). Changes in the properties of CPCs with time may involve loss of self-renewing capacity, increased symmetric division with formation of daughter committed cells, partial depletion of the primitive pool, biased differentiation to the fibroblast fate, impaired ability to migrate, and forced entry into an irreversible quiescent state. Telomere shortening is a major variable of cellular senescence and organ aging, and support the notion that CPCs with critically shortened or dysfunctional telomeres contribute to myocardial aging and chronic heart failure. These defects constitute the critical variables that define the aging myopathy in humans. Importantly, a compartment of functionally competent human CPCs persists in the decompensated heart pointing to stem cell therapy as a novel form of treatment for the aging myopathy.


Assuntos
Coração/crescimento & desenvolvimento , Mioblastos Cardíacos/fisiologia , Miócitos Cardíacos/fisiologia , Animais , Senescência Celular , Coração/fisiologia , Humanos , Mioblastos Cardíacos/citologia , Mioblastos Cardíacos/metabolismo , Miócitos Cardíacos/citologia , Miócitos Cardíacos/metabolismo
15.
Philos Trans R Soc Lond B Biol Sci ; 370(1680): 20140373, 2015 Oct 19.
Artigo em Inglês | MEDLINE | ID: mdl-26416682

RESUMO

Stem cell-based therapy is currently tested in several trials of chronic heart failure. The main question is to determine how its implementation could be extended to common clinical practice. To fill this gap, it is critical to first validate the hypothesis that the grafted stem cells primarily act by harnessing endogenous repair pathways. The confirmation of this mechanism would have three major clinically relevant consequences: (i) the use of cardiac-committed cells, since even though cells primarily act in a paracrine manner, such a phenotype seems the most functionally effective; (ii) the optimization of early cell retention, rather than of sustained cell survival, so that the cells reside in the target tissue long enough to deliver the factors underpinning their action; and (iii) the reliance on allogeneic cells, the expected rejection of which should only have to be delayed since a permanent engraftment would no longer be the objective. One step further, the long-term objective of cell therapy could be to use the cells exclusively for producing factors and then to only administer them to the patient. The production process would then be closer to that of a biological pharmaceutic, thereby facilitating an extended clinical use.


Assuntos
Insuficiência Cardíaca/terapia , Transplante de Células-Tronco , Técnicas de Cultura de Células , Linhagem da Célula , Sobrevivência Celular , Humanos , Modelos Cardiovasculares , Mioblastos Cardíacos/fisiologia , Mioblastos Cardíacos/transplante , Transplante Autólogo , Transplante Homólogo
16.
Circ J ; 79(7): 1422-30, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26073608

RESUMO

Over the past 2 decades, cardiac regeneration has evolved from an exotic fringe of cardiovascular biology to the forefront of molecular, genetic, epigenetic, translational, and clinical investigations. The unmet patient need is the paucity of self-repair following infarction. Robust regeneration seen in models such as zebrafish and newborn mice has inspired the field, along with encouragement from modern methods that make even low levels of restorative growth discernible, changing the scientific and technical landscape for effective counter-measures. Approaches under study to augment cardiac repair complement each other, and encompass grafting cells of diverse kinds, restarting the cell cycle in post-mitotic ventricular myocytes, reprogramming non-myocytes, and exploiting the dormant progenitor/stem cells that lurk within the adult heart. The latter are the emphasis of the present review. Cardiac-resident stem cells (CSC) can be harvested from heart tissue, expanded, and delivered to the myocardium as a therapeutic product, whose benefits may be hoped to surpass those achieved in human trials of bone marrow. However, important questions are prompted by such cells' discovery. How do they benefit recipient hearts? Do they contribute, measurably, as an endogenous population, to self-repair? Even if "no," might CSCs be targets for activation in situ by growth factors and other developmental catalysts? And, what combination of distinguishing markers best demarcates the cells with robust clonal growth and cardiogenic potential?


Assuntos
Células-Tronco Adultas/citologia , Mioblastos Cardíacos/citologia , Adulto , Células-Tronco Adultas/fisiologia , Células-Tronco Adultas/transplante , Animais , Biomarcadores , Diferenciação Celular , Linhagem da Célula , Terapia Baseada em Transplante de Células e Tecidos , Perfilação da Expressão Gênica , Coração/fisiologia , Humanos , Camundongos , Modelos Animais , Modelos Cardiovasculares , Mioblastos Cardíacos/fisiologia , Mioblastos Cardíacos/transplante , Regeneração/fisiologia , Peixe-Zebra
17.
Life Sci ; 132: 68-76, 2015 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-25916801

RESUMO

AIM: To study the effects of 2, 3, 5, 4'-tetrahydroxystilbene-2-O-ß-d-glucoside (THSG) on proliferation of rat cardiac stem cells (CSCs) in vitro. MATERIALS AND METHODS: C-kit(+) cells were isolated from neonatal (1 day old) Sprague-Dawley rats by using flow cytometry. Optimal THSG treatment times and doses for growth of CSCs were analyzed. CSCs were treated with various THSG doses (0, 1, 10, and 100 µM) for 12h. RESULTS: Sorted c-kit(+) cells exhibited self-renewing and clonogenic capabilities. Cell Counting Kit (CCK-8) and Proliferating Cell Nuclear Antigen (PCNA) ELISA test positive cells were significantly increased in THSG-treated groups compared with untreated controls. The percentage of S-phase cells also increased after THSG treatment. Moreover, we show that some c-kit(+) cells spontaneously express vascular endothelial growth factor (VEGF), T-box transcription factor (Tbx5), hyperpolarization-activated cyclic nucleotide-gated 2 (HCN2), hyperpolarization-activated cyclic nucleotide gated 4 (HCN4), alpha myosin heavy chain (αMHC), and beta myosin heavy chain (ßMHC) mRNA, and stem cell antigen 1 (Sca-1), cardiac troponin-I, GATA-4, Nkx2.5, and connexin 43 protein were also assessed in CSCs. However, their expression was significantly increased with THSG treatment when compared to untreated controls. CONCLUSION: THSG can increase proliferation of rat CSCs in vitro and thus, shows promise as a potential treatment strategy for stimulating endogenous stem cells to help repair the injured heart after myocardial infarction in patients.


Assuntos
Proliferação de Células/efeitos dos fármacos , Glucosídeos/farmacologia , Mioblastos Cardíacos/fisiologia , Miocárdio/citologia , Estilbenos/farmacologia , Análise de Variância , Animais , Western Blotting , Células Cultivadas , Primers do DNA/genética , Relação Dose-Resposta a Droga , Avaliação Pré-Clínica de Medicamentos , Ensaio de Imunoadsorção Enzimática , Citometria de Fluxo , Perfilação da Expressão Gênica , Técnicas In Vitro , Mioblastos Cardíacos/efeitos dos fármacos , Antígeno Nuclear de Célula em Proliferação/metabolismo , Ratos , Ratos Sprague-Dawley , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fator de Células-Tronco/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo
18.
EMBO J ; 34(6): 759-77, 2015 Mar 12.
Artigo em Inglês | MEDLINE | ID: mdl-25564442

RESUMO

Scl/Tal1 confers hemogenic competence and prevents ectopic cardiomyogenesis in embryonic endothelium by unknown mechanisms. We discovered that Scl binds to hematopoietic and cardiac enhancers that become epigenetically primed in multipotent cardiovascular mesoderm, to regulate the divergence of hematopoietic and cardiac lineages. Scl does not act as a pioneer factor but rather exploits a pre-established epigenetic landscape. As the blood lineage emerges, Scl binding and active epigenetic modifications are sustained in hematopoietic enhancers, whereas cardiac enhancers are decommissioned by removal of active epigenetic marks. Our data suggest that, rather than recruiting corepressors to enhancers, Scl prevents ectopic cardiogenesis by occupying enhancers that cardiac factors, such as Gata4 and Hand1, use for gene activation. Although hematopoietic Gata factors bind with Scl to both activated and repressed genes, they are dispensable for cardiac repression, but necessary for activating genes that enable hematopoietic stem/progenitor cell development. These results suggest that a unique subset of enhancers in lineage-specific genes that are accessible for regulators of opposing fates during the time of the fate decision provide a platform where the divergence of mutually exclusive fates is orchestrated.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Diferenciação Celular/fisiologia , Elementos Facilitadores Genéticos/fisiologia , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Células-Tronco Hematopoéticas/citologia , Mesoderma/embriologia , Mioblastos Cardíacos/citologia , Proteínas Proto-Oncogênicas/metabolismo , Células Cultivadas , Imunoprecipitação da Cromatina , Perfilação da Expressão Gênica , Biblioteca Gênica , Células-Tronco Hematopoéticas/fisiologia , Humanos , Mesoderma/metabolismo , Análise em Microsséries , Modelos Biológicos , Dados de Sequência Molecular , Mioblastos Cardíacos/fisiologia , Análise de Sequência de RNA , Proteína 1 de Leucemia Linfocítica Aguda de Células T
19.
Mol Cell Biochem ; 401(1-2): 61-70, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25481685

RESUMO

Sulforaphane is a naturally occurring isothiocyanate capable of stimulating cellular antioxidant defenses and inducing phase 2 detoxifying enzymes, which can protect cells against oxidative damage. Oxidative stress and apoptosis are intimately involved in the pathophysiology of cardiac diseases. Although sulforaphane is known for its anticancer benefits, its role in cardiac cells is just emerging. The aim of the present study was to investigate whether sulforaphane can modulate oxidative stress, apoptosis, and correlate with PGC-1α, a transcriptional cofactor involved in energy metabolism. H9c2 cardiac myoblasts were incubated with R-sulforaphane 5 µmol/L for 24 h. Cell viability, ANP gene expression, oxidative stress and apoptosis markers, and protein expression of PGC-1α were studied. In cells treated with sulforaphane, cellular viability increased (12 %) and ANP gene expression decreased (46 %) compared to control cells. Moreover, sulforaphane induced a significant increase in superoxide dismutase (103 %), catalase (101 %), and glutathione S-transferase (72 %) activity, reduced reactive oxygen species levels (15 %) and lipid peroxidation (65 %), as well as stimulated the expression of the cytoprotective enzyme heme oxygenase-1 (4-fold). Sulforaphane also promoted an increase in the expression of the anti-apoptotic protein Bcl-2 (60 %), decreasing the Bax/Bcl-2 ratio. Active Caspase 3\7 and p-JNK/JNK were also reduced by sulforaphane, suggesting a reduction in apoptotic signaling. This was associated with an increased protein expression of PGC-1α (42 %). These results suggest that sulforaphane offers cytoprotection to cardiac cells by activating PGC1-α, reducing oxidative stress, and decreasing apoptosis signaling.


Assuntos
Antioxidantes/farmacologia , Isotiocianatos/farmacologia , Mioblastos Cardíacos/efeitos dos fármacos , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Animais , Apoptose , Fator Natriurético Atrial/genética , Fator Natriurético Atrial/metabolismo , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Regulação da Expressão Gênica/efeitos dos fármacos , Mioblastos Cardíacos/fisiologia , Estresse Oxidativo/efeitos dos fármacos , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo , Ratos , Transdução de Sinais/efeitos dos fármacos , Sulfóxidos
20.
Cardiovasc Res ; 105(1): 44-54, 2015 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-25362681

RESUMO

AIMS: Our previous study found that A83-01, a small molecule type 1 TGFß receptor inhibitor, could induce proliferation of postnatal Nkx2.5(+) cardiomyoblasts in vitro and enhance their cardiomyogenic differentiation. The present study addresses whether A83-01 treatment in vivo could increase cardiomyogenesis and improve cardiac function after myocardial infarction through an Nkx2.5(+) cardiomyoblast-dependent process. METHODS AND RESULTS: To determine the effect of A83-01 on the number of Nkx2.5(+) cardiomyoblasts in the heart after myocardial injury, we treated transgenic Nkx2.5 enhancer-GFP reporter mice for 7 days with either A83-01 or DMSO and measured the number of GFP(+) cardiomyoblasts in the heart at 1 week after injury by flow cytometry. To determine the degree of new cardiomyocyte formation after myocardial injury and the effect of A83-01 in this process, we employed inducible Nkx2.5 enhancer-Cre transgenic mice to lineage label postnatal Nkx2.5(+) cardiomyoblasts and their differentiated progenies after myocardial injury. We also examined the cardiac function of each animal by intracardiac haemodynamic measurements. We found that A83-01 treatment significantly increased the number of Nkx2.5(+) cardiomyoblasts at baseline and after myocardial injury, resulting in an increase in newly formed cardiomyocytes. Finally, we showed that A83-01 treatment significantly improved ventricular elastance and stroke work, leading to improved contractility after injury. CONCLUSION: Pharmacological inhibition of TGFß signalling improved cardiac function in injured mice and promoted the expansion and cardiomyogenic differentiation of Nkx2.5(+) cardiomyoblasts. Direct modulation of resident cardiomyoblasts in vivo may be a promising strategy to enhance therapeutic cardiac regeneration.


Assuntos
Proteínas de Homeodomínio/metabolismo , Mioblastos Cardíacos/efeitos dos fármacos , Mioblastos Cardíacos/fisiologia , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Pirazóis/farmacologia , Receptores de Fatores de Crescimento Transformadores beta/antagonistas & inibidores , Regeneração/efeitos dos fármacos , Regeneração/fisiologia , Tiossemicarbazonas/farmacologia , Fatores de Transcrição/metabolismo , Animais , Diferenciação Celular , Modelos Animais de Doenças , Feminino , Técnicas de Silenciamento de Genes , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Proteína Homeobox Nkx-2.5 , Proteínas de Homeodomínio/antagonistas & inibidores , Proteínas de Homeodomínio/genética , Camundongos , Camundongos Transgênicos , Mioblastos Cardíacos/citologia , Infarto do Miocárdio/tratamento farmacológico , Infarto do Miocárdio/patologia , Infarto do Miocárdio/fisiopatologia , Gravidez , RNA Interferente Pequeno/genética , Receptor do Fator de Crescimento Transformador beta Tipo I , Fatores de Transcrição/antagonistas & inibidores , Fatores de Transcrição/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA