Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 75
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
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.
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
3.
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
4.
Circ Res ; 114(1): 41-55, 2014 Jan 03.
Artigo em Inglês | MEDLINE | ID: mdl-24170267

RESUMO

RATIONALE: Hypoxia favors stem cell quiescence, whereas normoxia is required for stem cell activation, but whether cardiac stem cell (CSC) function is regulated by the hypoxic/normoxic state of the cell is currently unknown. OBJECTIVE: A balance between hypoxic and normoxic CSCs may be present in the young heart, although this homeostatic control may be disrupted with aging. Defects in tissue oxygenation occur in the old myocardium, and this phenomenon may expand the pool of hypoxic CSCs, which are no longer involved in myocyte renewal. METHODS AND RESULTS: Here, we show that the senescent heart is characterized by an increased number of quiescent CSCs with intact telomeres that cannot re-enter the cell cycle and form a differentiated progeny. Conversely, myocyte replacement is controlled only by frequently dividing CSCs with shortened telomeres; these CSCs generate a myocyte population that is chronologically young but phenotypically old. Telomere dysfunction dictates their actual age and mechanical behavior. However, the residual subset of quiescent young CSCs can be stimulated in situ by stem cell factor reversing the aging myopathy. CONCLUSIONS: Our findings support the notion that strategies targeting CSC activation and growth interfere with the manifestations of myocardial aging in an animal model. Although caution has to be exercised in the translation of animal studies to human beings, our data strongly suggest that a pool of functionally competent CSCs persists in the senescent heart and that this stem cell compartment can promote myocyte regeneration effectively, partly correcting the aging myopathy.


Assuntos
Envelhecimento/efeitos dos fármacos , Cardiomiopatias/metabolismo , Hipóxia/metabolismo , Mioblastos Cardíacos/metabolismo , Proteínas Proto-Oncogênicas c-kit/metabolismo , Fator de Células-Tronco/farmacologia , Nicho de Células-Tronco , Envelhecimento/metabolismo , Animais , Cardiomiopatias/tratamento farmacológico , Cardiomiopatias/patologia , Ciclo Celular , Linhagem da Célula , Proliferação de Células , Senescência Celular/efeitos dos fármacos , Hipóxia/patologia , Camundongos , Camundongos Endogâmicos C57BL , Mioblastos Cardíacos/efeitos dos fármacos , Mioblastos Cardíacos/fisiologia , Miocárdio/metabolismo , Miocárdio/patologia , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/fisiologia , Fator de Células-Tronco/uso terapêutico , Homeostase do Telômero
5.
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
6.
Dev Biol ; 396(2): 169-82, 2014 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-25224224

RESUMO

The Drosophila heart is a linear organ formed by the movement of bilaterally specified progenitor cells to the midline and adherence of contralateral heart cells. This movement occurs through the attachment of heart cells to the overlying ectoderm which is undergoing dorsal closure. Therefore heart cells are thought to move to the midline passively. Through live imaging experiments and analysis of mutants that affect the speed of dorsal closure we show that heart cells in Drosophila are autonomously migratory and part of their movement to the midline is independent of the ectoderm. This means that heart formation in flies is more similar to that in vertebrates than previously thought. We also show that defects in dorsal closure can result in failure of the amnioserosa to properly degenerate, which can physically hinder joining of contralateral heart cells leading to a broken heart phenotype.


Assuntos
Movimento Celular/fisiologia , Proteínas de Drosophila/genética , Drosophila/embriologia , Coração/embriologia , Proteínas de Membrana/genética , Mioblastos Cardíacos/fisiologia , Organogênese/fisiologia , Fosfatidato Fosfatase/genética , Animais , Imuno-Histoquímica , Microscopia de Fluorescência
7.
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
8.
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
9.
Proc Natl Acad Sci U S A ; 109(32): 13016-21, 2012 Aug 07.
Artigo em Inglês | MEDLINE | ID: mdl-22826236

RESUMO

Unique insights for the reprograming of cell lineages have come from embryonic development in the ascidian Ciona, which is dependent upon the transcription factors Ci-ets1/2 and Ci-mesp to generate cardiac progenitors. We tested the idea that mammalian v-ets erythroblastosis virus E26 oncogene homolog 2 (ETS2) and mesoderm posterior (MESP) homolog may be used to convert human dermal fibroblasts into cardiac progenitors. Here we show that murine ETS2 has a critical role in directing cardiac progenitors during cardiopoiesis in embryonic stem cells. We then use lentivirus-mediated forced expression of human ETS2 to convert normal human dermal fibroblasts into replicative cells expressing the cardiac mesoderm marker KDR(+). However, although neither ETS2 nor the purported cardiac master regulator MESP1 can by themselves generate cardiac progenitors de novo from fibroblasts, forced coexpression of ETS2 and MESP1 or cell treatment with purified proteins reprograms fibroblasts into cardiac progenitors, as shown by the de novo appearance of core cardiac transcription factors, Ca(2+) transients, and sarcomeres. Our data indicate that ETS2 and MESP1 play important roles in a genetic network that governs cardiopoiesis.


Assuntos
Transdiferenciação Celular/fisiologia , Fibroblastos/citologia , Mioblastos Cardíacos/citologia , Proteína Proto-Oncogênica c-ets-2/metabolismo , Pele/citologia , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Western Blotting , Transdiferenciação Celular/genética , Citometria de Fluxo , Imunofluorescência , Técnicas de Inativação de Genes , Humanos , Camundongos , Mioblastos Cardíacos/fisiologia , Reação em Cadeia da Polimerase , Proteína Proto-Oncogênica c-ets-2/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa
10.
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
11.
Dev Biol ; 373(1): 163-75, 2013 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-23103584

RESUMO

To evaluate potential roles of nitric oxide (NO) in the regulation of the endothelial lineage and neovascular processes (vasculogenesis and angiogenesis) we evaluated endothelial nitric oxide synthase (eNOS) and phosphorylated eNOS (p-eNOS) expression in 7.2-8.5 days post-coitum (dpc) mouse embryos. Analysis revealed that p-eNOS((S1177)) but not P-eNOS((S617)) or P-eNOS((T495)) was expressed in a subpopulation of angioblasts (TAL-1(+)/Flk-1(+)/CD31(-)/CD34(-)/VE-Cadherin(-)) at 7.2 dpc. A role of the VEGF/Akt1/eNOS signaling pathway in the regulation of the endothelial cell (EC) lineage was suggested by the strong correlation observed between cell division and p-eNOS((S1177)) expression in both angioblasts and embryonic endothelial cells (EECs, TAL-1(+)/Flk-1(+)/CD31(+)/CD34(+)/VE-Cadherin(+)). Our studies using Akt1 null mouse embryos show a reduction in p-eNOS((S1177)) expression in angioblast and EECs that is correlated with a decrease in endothelial cell proliferation and results in changes in VEGF-induced vascular patterning. Further, we show that VEGF-mediated cell proliferation in Flk-1(+) cells in allantoic cultures is decreased by pharmacological inhibitors of the VEGF/Akt1/eNOS signaling pathways. Taken together, our findings suggest that VEGF-mediated eNOS phosphorylation on Ser1177 regulates angioblast and EEC division, which underlies the formation of blood vessels and vascular networks.


Assuntos
Proliferação de Células , Células Endoteliais/fisiologia , Mioblastos Cardíacos/fisiologia , Neovascularização Fisiológica/fisiologia , Óxido Nítrico Sintase Tipo III/metabolismo , Transdução de Sinais/fisiologia , Fator A de Crescimento do Endotélio Vascular/metabolismo , Alantoide/citologia , Animais , Linhagem Celular , Linhagem da Célula/fisiologia , Células Endoteliais/metabolismo , Citometria de Fluxo , Imuno-Histoquímica , Camundongos , Microscopia de Fluorescência , Mioblastos Cardíacos/metabolismo , Fosforilação , Transdução de Sinais/genética
12.
Int J Sports Med ; 35(3): 181-90, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23900898

RESUMO

Cardiovascular diseases (CVD) are a global epidemic in developed countries. Cumulative evidence suggests that myocyte formation is preserved during postnatal life, in adulthood or senescence, suggesting the existence of a growth reserve of the heart throughout lifespan. Several medical therapeutic approaches to CVD have considerably improved the clinical outcome for patients. Intense interest has been focused on regenerative medicine as an emerging strategy for CVD. Cellular therapeutic approaches have been proposed for enhancing survival and propagation of stem cells in myocardium, leading to cardiac cellular repair. Strong epidemiological and clinical data exists concerning the impact of regular physical exercise on cardiovascular health. Several mechanisms of acute and chronic exercise-induced cardiovascular adaptations to exercise have been presented, considering primary and secondary prevention of CVD. In this context, exercise-related improvements in the function and regeneration of the cardiovascular system may be associated with the exercise-induced activation, mobilization, differentiation, and homing of stem and progenitor cells. In this review several topics will be addressed concerning the relation between exercise, recruitment and biological activity of blood-circulating progenitor cells and resident cardiac stem cells. We hypothesize that exercise-induced stem cell activation may enhance overall heart function and improve the efficacy of cardiac cellular therapeutic protocols.


Assuntos
Doenças Cardiovasculares/fisiopatologia , Doenças Cardiovasculares/terapia , Terapia Baseada em Transplante de Células e Tecidos , Exercício Físico/fisiologia , Coração/fisiopatologia , Mioblastos Cardíacos/fisiologia , Regeneração/fisiologia , Diferenciação Celular , Movimento Celular , Proliferação de Células , Humanos
13.
Eur Heart J ; 34(15): 1157-67, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22507976

RESUMO

AIMS: The study was designed to evaluate the mechanisms of cardiac regeneration after injury and to determine how to restore that capacity in aged individuals. The adult heart retains a small population of nascent cells that have myeloid, mesenchymal, and mesodermal capabilities, which play an essential role in the recovery of ventricular function after injury. In aged individuals, these cells are diminished and dysfunctional. We evaluated the derivation of some of these cardiac progenitors and a method to restore their number and function. METHODS AND RESULTS: We first demonstrated that aged mice have fewer progenitors in both the bone marrow (BM) and the myocardium, which correlated with the extent of cardiac dysfunction after injury. Bone marrow chimerism established in aged mice with young BM donors restored both myocardial progenitors and cardiac function, but neither was restored with aged BM donors. Cardiac micro-chimerism in aged mice was established with young BM cells, which restored cardiac function after injury, even with old peripheral BM cells. The young cardiac-resident BM-derived progenitor cells in the aged myocardium persisted for at least a year, and after myocardial infarction they actively proliferated and enhanced cardiac repair through paracrine mechanisms. CONCLUSION: Bone marrow reconstitution with young BM cells in aged recipients restored progenitors in both the BM and, most importantly, the myocardium. The number and function of cardiac-resident BM-derived progenitor cells in the aged myocardium prior to injury was the major determinant for successful recovery of cardiac function. The aged heart was rejuvenated with young BM cells.


Assuntos
Células da Medula Óssea/fisiologia , Medula Óssea/fisiologia , Coração/fisiologia , Infarto do Miocárdio/patologia , Regeneração/fisiologia , Células-Tronco/fisiologia , Animais , Modelos Animais de Doenças , Ligadura , Camundongos , Camundongos Endogâmicos C57BL , Mioblastos Cardíacos/fisiologia , Miocárdio/patologia , Quimeras de Transplante , Disfunção Ventricular
14.
Bull Exp Biol Med ; 157(1): 143-5, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24909728

RESUMO

The use of triple systemic transplantation of cardiomyoblasts raised from the culture of allogenic bone marrow mesenchymal stem cells of a healthy donor according to the new medical technology licensed by Federal Service on Surveillance in Healthcare in the therapy of a patient with late radiation cardiomyopathy and radiation exudative pericarditis developed 45 years after radiation therapy for Hodgkin lymphoma. High efficiency of systemic transplantation of mesenchymal stem cells partially differentiated towards cardiomyocytes was demonstrated. The therapeutic effect persists for more than 2 years. Possible mechanisms of the therapeutic effect of this type of stem cells and the prospects of using cell therapy in the treatment of late radiation injuries of vital organs and tissues are discussed.


Assuntos
Cardiomiopatias/terapia , Transplante de Células-Tronco Mesenquimais , Mioblastos Cardíacos/transplante , Pericardite/terapia , Cardiomiopatias/diagnóstico por imagem , Cardiomiopatias/etiologia , Cardiomiopatias/patologia , Diferenciação Celular , Raios gama/efeitos adversos , Doença de Hodgkin/complicações , Doença de Hodgkin/diagnóstico por imagem , Doença de Hodgkin/patologia , Doença de Hodgkin/radioterapia , Humanos , Masculino , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/fisiologia , Pessoa de Meia-Idade , Mioblastos Cardíacos/citologia , Mioblastos Cardíacos/fisiologia , Pericardite/diagnóstico por imagem , Pericardite/etiologia , Pericardite/patologia , Transplante Homólogo , Resultado do Tratamento , Ultrassonografia
15.
Biochem Biophys Res Commun ; 440(2): 197-203, 2013 Oct 18.
Artigo em Inglês | MEDLINE | ID: mdl-23985350

RESUMO

A potent analog (HNG) of the endogenous peptide humanin protects against myocardial ischemia-reperfusion (MI-R) injury in vivo, decreasing infarct size and improving cardiac function. Since oxidative stress contributes to the damage from MI-R we tested the hypotheses that: (1) HNG offers cardioprotection through activation of antioxidant defense mechanisms leading to preservation of mitochondrial structure and that, (2) the activity of either of a pair of non-receptor tyrosine kinases, c-Abl and Arg is required for this protection. Rat cardiac myoblasts (H9C2 cells) were exposed to nanomolar concentrations of HNG and to hydrogen peroxide (H2O2). Cells treated with HNG in the presence of H2O2 demonstrated reduced intracellular reactive oxygen species (ROS), preserved mitochondrial membrane potential, ATP levels and mitochondrial structure. HNG induced activation of catalase and glutathione peroxidase (GPx) within 5 min and decreased the ratio of oxidized to reduced glutathione within 30 min. siRNA knockdown of both Abl and Arg, but neither alone, abolished the HNG-mediated reduction of ROS in myoblasts exposed to H2O2. These findings demonstrate an HNG-mediated, Abl- and Arg-dependent, rapid and sustained activation of critical cellular defense systems and attenuation of oxidative stress, providing mechanistic insights into the observed HNG-mediated cardioprotection in vivo.


Assuntos
Peptídeos e Proteínas de Sinalização Intracelular/farmacologia , Mioblastos Cardíacos/efeitos dos fármacos , Traumatismo por Reperfusão Miocárdica/prevenção & controle , Estresse Oxidativo/efeitos dos fármacos , Peptídeos/farmacologia , Animais , Antioxidantes/farmacologia , Catalase/metabolismo , Técnicas de Silenciamento de Genes , Glutationa Peroxidase/metabolismo , Peróxido de Hidrogênio/farmacologia , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Mioblastos Cardíacos/fisiologia , Fármacos Neuroprotetores/farmacologia , Proteínas Tirosina Quinases/fisiologia , Proteínas Proto-Oncogênicas c-abl/fisiologia , Ratos , Espécies Reativas de Oxigênio/metabolismo
16.
Proc Natl Acad Sci U S A ; 107(8): 3329-34, 2010 Feb 23.
Artigo em Inglês | MEDLINE | ID: mdl-19846783

RESUMO

Myocardial infarction resulting in irreversible loss of cardiomyocytes (CMs) remains a leading cause of heart failure. Although cell transplantation has modestly improved cardiac function, major challenges including increasing cell survival, engraftment, and functional integration with host tissue, remain. Embryonic stem cells (ESCs), which can be differentiated into cardiac progenitors (CPs) and CMs, represent a candidate cell source for cardiac cell therapy. However, it is not known what specific cell type or condition is the most appropriate for transplantation. This problem is exasperated by the lack of efficient and predictive strategies to screen the large numbers of parameters that may impact cell transplantation. We used a cardiac tissue model, engineered heart tissue (EHT), and quantitative molecular and electrophysiological analyses, to test transplantation conditions and specific cell populations for their potential to functionally integrate with the host tissue. In this study, we validated our analytical platform using contractile mouse neonatal CMs (nCMs) and noncontractile cardiac fibroblasts (cFBs), and screened for the integration potential of ESC-derived CMs and CPs (ESC-CMs and -CPs). Consistent with previous in vivo studies, cFB injection interfered with electrical signal propagation, whereas injected nCMs improved tissue function. Purified bioreactor-generated ESC-CMs exhibited a diminished capacity for electrophysiological integration; a result correlated with lower (compared with nCMs) connexin 43 expression. ESC-CPs, however, appeared able to appropriately mature and integrate into EHT, enhancing the amplitude of tissue contraction. Our results support the use of EHT as a model system to accelerate development of cardiac cell therapy strategies.


Assuntos
Insuficiência Cardíaca/cirurgia , Mioblastos Cardíacos/fisiologia , Contração Miocárdica , Células-Tronco Pluripotentes/citologia , Engenharia Tecidual/métodos , Animais , Reatores Biológicos , Diferenciação Celular , Conexina 43/biossíntese , Fenômenos Eletrofisiológicos , Fibroblastos/fisiologia , Insuficiência Cardíaca/etiologia , Camundongos , Camundongos Transgênicos , Mioblastos Cardíacos/metabolismo , Mioblastos Cardíacos/transplante , Infarto do Miocárdio/complicações , Infarto do Miocárdio/fisiopatologia , Infarto do Miocárdio/cirurgia , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/fisiologia , Ratos , Ratos Sprague-Dawley
17.
Dev Dyn ; 241(2): 284-93, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22184055

RESUMO

BACKGROUND: The cardiac progenitor cells for the outflow tract (OFT) reside in the visceral mesoderm and mesodermal core of the pharyngeal region, which are defined as the secondary and anterior heart fields (SHF and AHF), respectively. RESULTS: Using chick embryos, we injected fluorescent-dye into the SHF or AHF at stage 14, and the destinations of the labeled cells were examined at stage 31. Labeled cells from the right SHF were found in the myocardium on the left dorsal side of the OFT, and cells from the left SHF were detected on the right ventral side of the OFT. Labeled cells from the right and left AHF migrated to regions of the ventral wall of the OFT close to the aortic and pulmonary valves, respectively. CONCLUSION: These observations indicate that myocardial progenitors from the SHF and AHF contribute to distinct conotruncal regions and that cells from the SHF migrate rotationally while cells from the AHF migrate in a non-rotational manner.


Assuntos
Movimento Celular , Mioblastos Cardíacos/fisiologia , Miocárdio , Faringe/embriologia , Animais , Embrião de Galinha , Cardiopatias Congênitas/embriologia , Faringe/citologia
18.
J Cell Mol Med ; 16(5): 961-5, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22212626

RESUMO

While recent data have supported the capacity for a neonatal heart to undergo cardiomyogenesis, it is unclear whether these new cardiomyocytes arise from an immature cardiomyoblast population or from the division of mature cardiomyocytes. By following the expression of enhanced Green Fluorescent Protein (eGFP) in an Nkx2.5 enhancer-eGFP transgenic mice, we have identified a population of immature cells that can undergo cardiomyogenic as well as smooth muscle cell differentiation in the neonatal heart. Here, we examined growth factors and small molecule regulators that potentially regulate the proliferation and cardiomyogenic versus smooth muscle cell differentiation of neonatal Nkx2.5-GFP (+) cells in vitro. We found that A83-01 (A83), an inhibitor of TGF-ßRI, was able to induce an expansion of neonatal Nkx2.5-eGFP (+) cells. In addition, the ability of A83 to expand eGFP (+) cells in culture was dependent on signalling from the mitogen-activated protein kinase kinase (MEK) as treatment with a MEK inhibitor, PD0325901, abolished this effect. On the other hand, activation of neonatal Nkx2.5-eGFP (+) cells with TGF-ß1, but not activin A nor BMP2, led to smooth muscle cell differentiation, an effect that can be reversed by treatment with A83. In summary, small molecule inhibition of TGF-ß signalling may be a promising strategy to induce the expansion of a rare population of postnatal cardiomyoblasts.


Assuntos
Proliferação de Células/efeitos dos fármacos , Proteínas de Homeodomínio/metabolismo , Desenvolvimento Muscular/fisiologia , Mioblastos Cardíacos/efeitos dos fármacos , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Pirazóis/farmacologia , Receptores de Fatores de Crescimento Transformadores beta/antagonistas & inibidores , Tiocarbamatos/farmacologia , Fatores de Transcrição/metabolismo , Ativinas/antagonistas & inibidores , Ativinas/farmacologia , Animais , Benzamidas/farmacologia , Proteína Morfogenética Óssea 2/antagonistas & inibidores , Proteína Morfogenética Óssea 2/farmacologia , Células Cultivadas , Difenilamina/análogos & derivados , Difenilamina/farmacologia , Inibidores Enzimáticos/farmacologia , Proteína Homeobox Nkx-2.5 , Proteínas de Homeodomínio/genética , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/fisiologia , Camundongos , Camundongos Transgênicos , Quinases de Proteína Quinase Ativadas por Mitógeno/antagonistas & inibidores , Desenvolvimento Muscular/efeitos dos fármacos , Mioblastos Cardíacos/fisiologia , Receptor do Fator de Crescimento Transformador beta Tipo I , Tiossemicarbazonas , Fatores de Transcrição/genética , Fator de Crescimento Transformador beta1/farmacologia
19.
BMC Dev Biol ; 12: 8, 2012 Feb 21.
Artigo em Inglês | MEDLINE | ID: mdl-22353787

RESUMO

BACKGROUND: The formation of a tubular organ, such as the heart, requires the communication of positional and polarity signals between migratory cells. Key to this process is the establishment of a new luminal domain on the cell surface, generally from the apical domain of a migratory cell. This domain will also acquire basal properties, as it will produce a luminal extracellular matrix. Integrin receptors are the primary means of cell adhesion and adhesion signaling with the extracellular matrix. Here we characterise the requirement of Integrins in a genetic model of vasculogenesis, the formation of the heart in Drosophila. RESULTS: As with vertebrates, the Drosophila heart arises from lateral mesoderm that migrates medially to meet their contralateral partners, to then assemble a midline vessel. During migration, Integrins are among the first proteins restricted to the presumptive luminal domain of cardioblasts. Integrins are required for normal levels of leading edge membrane motility. Apical accumulation of Integrins is enhanced by Robo, and reciprocally, apicalisation of luminal factors like Slit and Robo requires Integrin function. Integrins may provide a template for the formation of a lumen by stabilising lumen factors like Robo. Subsequent to migration, Integrin is required for normal cardioblast alignment and lumen formation. This phenotype is most readily modified by other mutations that affect adhesion, such as Talin and extracellular matrix ligands. CONCLUSION: Our findings reveal an instructive role for Integrins in communicating polarising information to cells during migration, and during transition to an epithelial tube structure.


Assuntos
Polaridade Celular , Proteínas de Drosophila/fisiologia , Drosophila melanogaster/embriologia , Coração/embriologia , Cadeias alfa de Integrinas/fisiologia , Mioblastos Cardíacos/fisiologia , Miocárdio/citologia , Animais , Movimento Celular , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/citologia , Drosophila melanogaster/metabolismo , Proteínas da Matriz Extracelular/genética , Proteínas da Matriz Extracelular/metabolismo , Redes Reguladoras de Genes , Cadeias alfa de Integrinas/metabolismo , Mioblastos Cardíacos/metabolismo , Mioblastos Cardíacos/ultraestrutura , Miocárdio/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Transporte Proteico , Receptores Imunológicos/metabolismo , Transdução de Sinais/genética , Proteínas Roundabout
20.
Circulation ; 123(4): 364-73, 2011 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-21242485

RESUMO

BACKGROUND: Human cardiac progenitor cells (hCPCs) may promote myocardial regeneration in adult ischemic myocardium. The regenerative capacity of hCPCs in young patients with nonischemic congenital heart defects for potential use in congenital heart defect repair warrants exploration. METHODS AND RESULTS: Human right atrial specimens were obtained during routine congenital cardiac surgery across 3 groups: neonates (age, <30 days), infants (age, 1 month to 2 years), and children (age, >2 to ≤13 years). C-kit(+) hCPCs were 3-fold higher in neonates than in children >2 years of age. hCPC proliferation was greatest during the neonatal period as evidenced by c-kit(+) Ki67(+) expression but decreased with age. hCPC differentiation capacity was also greatest in neonatal right atrium as evidenced by c-kit(+), NKX2-5(+), NOTCH1(+), and NUMB(+) expression. Despite the age-dependent decline in resident hCPCs, we isolated and expanded right atrium-derived CPCs from all patients (n=103) across all ages and diagnoses using the cardiosphere method. Intact cardiospheres contained a mix of heart-derived cell subpopulations that included cardiac progenitor cells expressing c-kit(+), Islet-1, and supporting cells. The number of c-kit(+)-expressing cells was highest in human cardiosphere-derived cells (hCDCs) grown from neonatal and infant right atrium. Furthermore, hCDCs could differentiate into diverse cardiovascular lineages by in vitro differentiation assays. Transplanted hCDCs promoted greater myocardial regeneration and functional improvement in infarcted myocardium than transplanted cardiac fibroblasts. CONCLUSIONS: Resident hCPCs are most abundant in the neonatal period and rapidly decrease over time. hCDCs can be reproducibly isolated and expanded from young human myocardial samples regardless of age or diagnosis. hCPCs are functional and have potential in congenital cardiac repair.


Assuntos
Cardiopatias Congênitas/cirurgia , Mioblastos Cardíacos/fisiologia , Mioblastos Cardíacos/transplante , Adolescente , Fatores Etários , Animais , Diferenciação Celular , Proliferação de Células , Criança , Pré-Escolar , Ensaios Clínicos como Assunto , Proteína Homeobox Nkx-2.5 , Proteínas de Homeodomínio/metabolismo , Humanos , Lactente , Recém-Nascido , Antígeno Ki-67/metabolismo , Masculino , Proteínas de Membrana/metabolismo , Mioblastos Cardíacos/citologia , Proteínas do Tecido Nervoso/metabolismo , Proteínas Proto-Oncogênicas c-kit/metabolismo , Ratos , Ratos Nus , Receptor Notch1/metabolismo , Fatores de Transcrição/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA