Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
PLoS One ; 10(2): e0116600, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25692855

RESUMO

In the developing heart, cardiomyocytes undergo terminal differentiation during a critical window around birth. Hypoxia is a major stress to preterm infants, yet its effect on the development and maturation of the heart remains unknown. We tested the hypothesis in a rat model that newborn anoxia accelerates cardiomyocyte terminal differentiation and results in reduced cardiomyocyte endowment in the developing heart via an endothelin-1-dependent mechanism. Newborn rats were exposed to anoxia twice daily from postnatal day 1 to 3, and hearts were isolated and studied at postnatal day 4 (P4), 7 (P7), and 14 (P14). Anoxia significantly increased HIF-1α protein expression and pre-proET-1 mRNA abundance in P4 neonatal hearts. Cardiomyocyte proliferation was significantly decreased by anoxia in P4 and P7, resulting in a significant reduction of cardiomyocyte number per heart weight in the P14 neonates. Furthermore, the expression of cyclin D2 was significantly decreased due to anoxia, while p27 expression was increased. Anoxia has no significant effect on cardiomyocyte binucleation or myocyte size. Consistently, prenatal hypoxia significantly decreased cardiomyocyte proliferation but had no effect on binucleation in the fetal heart. Newborn administration of PD156707, an ETA-receptor antagonist, significantly increased cardiomyocyte proliferation at P4 and cell size at P7, resulting in an increase in the heart to body weight ratio in P7 neonates. In addition, PD156707 abrogated the anoxia-mediated effects. The results suggest that hypoxia and anoxia via activation of endothelin-1 at the critical window of heart development inhibits cardiomyocyte proliferation and decreases myocyte endowment in the developing heart, which may negatively impact cardiac function later in life.


Assuntos
Endotelina-1/metabolismo , Coração Fetal/citologia , Coração Fetal/embriologia , Miócitos Cardíacos/citologia , Animais , Animais Recém-Nascidos , Peso Corporal/efeitos dos fármacos , Contagem de Células , Hipóxia Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Tamanho Celular/efeitos dos fármacos , Dioxóis/farmacologia , Antagonistas do Receptor de Endotelina A/farmacologia , Endotelina-1/genética , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Subunidade alfa do Fator 1 Induzível por Hipóxia , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/metabolismo , Tamanho do Órgão/efeitos dos fármacos , Gravidez , Antígeno Nuclear de Célula em Proliferação/metabolismo , Precursores de Proteínas/genética , Precursores de Proteínas/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley , Receptor de Endotelina A/metabolismo
2.
Drug Discov Today ; 20(2): 223-35, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25461956

RESUMO

Recent studies provide evidence that ischemic preconditioning (IP) and ischemia/reperfusion (IR) injury lead to altered expression of microRNAs (miRNAs) that affect the survival and recovery of cardiomyocytes. These endogenous ∼22-nucleotide noncoding RNAs negatively regulate gene expression via degradation and translational inhibition of their target mRNAs. miRNAs are involved in differentiation, proliferation, electrical conduction, angiogenesis and apoptosis. These pathways can lead to physiological and pathological adaptations. This review intends to explore several facets of miRNA expression and the underlying mechanisms involved in IR injury, as well as IP as a cardioprotective strategy. In addition, we will investigate miRNA interaction with the renin-angiotensin system and the potential use of miRNAs in developing sensitive biomarkers for cardiovascular disease.


Assuntos
MicroRNAs , Isquemia Miocárdica/genética , Animais , Biomarcadores/metabolismo , Humanos , Precondicionamento Isquêmico Miocárdico , MicroRNAs/genética , MicroRNAs/metabolismo , Sistema Renina-Angiotensina
3.
Int J Med Sci ; 11(4): 373-80, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24578615

RESUMO

AIMS: Hypoxia is a major stress on fetal development and leads to induction of endothelin-1 (ET-1) expression. We tested the hypothesis that ET-1 stimulates the terminal differentiation of cardiomyocytes from mononucleate to binucleate in the developing heart. METHODS AND RESULTS: Hypoxia (10.5% O2) treatment of pregnant rats from day 15 to day 21 resulted in a significant increase in prepro-ET-1 mRNA expression in fetal hearts. ET-1 ex vivo treatment of fetal rat cardiomyocytes increased percent binucleate cells and decreased Ki-67 expression, a marker for proliferation, under both control and hypoxic conditions. Hypoxia alone decreased Ki-67 expression and in conjunction with ET-1 treatment decreased cardiomyocyte size. PD145065, a non-selective ET-receptor antagonist, blocked the changes in binucleation and proliferation caused by ET-1. DNA methylation in fetal cardiomyocytes was significantly increased with ET-1 treatment, which was blocked by 5-aza-2'-deoxycytidine, a DNA methylation inhibitor. In addition, 5-aza-2'-deoxycytidine treatment abrogated the increase in binucleation and decrease in proliferation induced by ET-1. CONCLUSIONS: Hypoxic stress and synthesis of ET-1 increases DNA methylation and promotes terminal differentiation of cardiomyocytes in the developing heart. This premature exit of the cell cycle may lead to a reduced cardiomyocyte endowment in the heart and have a negative impact on cardiac function.


Assuntos
Metilação de DNA/fisiologia , Endotelina-1/metabolismo , Miócitos Cardíacos/citologia , Miócitos Cardíacos/metabolismo , Animais , Metilação de DNA/genética , Endotelina-1/genética , Ensaio de Imunoadsorção Enzimática , Feminino , Hipóxia/metabolismo , Hipóxia/fisiopatologia , Imuno-Histoquímica , Gravidez , Reação em Cadeia da Polimerase em Tempo Real
4.
Drug Discov Today ; 19(5): 602-9, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24184431

RESUMO

Cardiomyocytes possess a unique ability to transition from mononucleate to the mature binucleate phenotype in late fetal development and around birth. Mononucleate cells are proliferative, whereas binucleate cells exit the cell cycle and no longer proliferate. This crucial period of terminal differentiation dictates cardiomyocyte endowment for life. Adverse early life events can influence development of the heart, affecting cardiomyocyte number and contributing to heart disease late in life. Although much is still unknown about the mechanisms underlying the binucleation process, many studies are focused on molecules involved in cell cycle regulation and cytokinesis as well as epigenetic modifications that can occur during this transition. Better understanding of these mechanisms could provide a basis for recovering the proliferative capacity of cardiomyocytes.


Assuntos
Diferenciação Celular/fisiologia , Proliferação de Células/fisiologia , Miócitos Cardíacos/fisiologia , Animais , Divisão Celular/fisiologia , Citocinese/fisiologia , Epigênese Genética/fisiologia , Desenvolvimento Fetal/fisiologia , Humanos
5.
Curr Vasc Pharmacol ; 11(5): 594-605, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-24063378

RESUMO

Endothelins are 21-amino acid peptides involved in vascular homeostasis. Three types of peptide have been identified, with endothelin-1 (ET-1) being the most potent vasoconstrictor currently known. Two endothelin receptor subtypes are found in various tissues, including the brain, heart, blood vessel, lung, and placenta. The ETA-receptor is associated with vasoconstriction in vascular smooth muscle. Conversely, the ETB-receptor can elicit a vasoconstrictor effect in vascular smooth muscle and a vasodilator effect via its action in endothelial cells. Both receptors play a key role in maintaining circulatory homeostasis and vascular function. Changes in ET-1 expression are found in various disease states, and overexpression of ET-1 is observed in hypertension and preeclampsia in pregnancy. Placental localization of ET-1 implies a key role in regulating the uteroplacental circulation. Additionally, ET-1 is important in the fetal circulation and is involved in the pulmonary circulation and closure of the ductus arteriosus after birth, as well as fetal growth constriction in utero. ET receptor antagonists and nitric oxide donors may provide therapeutic potential in treating conditions associated with overexpression of ET and hypertension.


Assuntos
Endotelina-1/metabolismo , Feto/irrigação sanguínea , Feto/fisiologia , Circulação Placentária/fisiologia , Animais , Endotelina-1/genética , Feminino , Feto/metabolismo , Humanos , Circulação Placentária/genética , Gravidez , Receptores de Endotelina/metabolismo
6.
Hypertension ; 62(3): 599-607, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23817493

RESUMO

Preeclampsia is a life-threatening pregnancy disorder. However, its pathogenesis remains unclear. We tested the hypothesis that gestational hypoxia induces preeclampsia-like symptoms via heightened endothelin-1 (ET-1) signaling. Time-dated pregnant and nonpregnant rats were divided into normoxic and hypoxic (10.5% O2 from the gestational day 6-21) groups. Chronic hypoxia had no significant effect on blood pressure or proteinuria in nonpregnant rats but significantly increased blood pressure on day 12 (systolic blood pressure, 111.7 ± 6.1 versus 138.5 ± 3.5 mm Hg; P=0.004) and day 20 (systolic blood pressure, 103.4 ± 4.6 versus 125.1 ± 6.1 mm Hg; P=0.02) in pregnant rats and urine protein (µg/µL)/creatinine (nmol/µL) ratio on day 20 (0.10 ± 0.01 versus 0.20 ± 0.04; P=0.04), as compared with the normoxic control group. This was accompanied with asymmetrical fetal growth restriction. Hypoxia resulted in impaired trophoblast invasion and uteroplacental vascular remodeling. In addition, plasma ET-1 levels, as well as the abundance of prepro-ET-1 mRNA, ET-1 type A receptor and angiotensin II type 1 receptor protein in the kidney and placenta were significantly increased in the chronic hypoxic group, as compared with the control animals. Treatment with the ET-1 type A receptor antagonist, BQ123, during the course of hypoxia exposure significantly attenuated the hypoxia-induced hypertension and other preeclampsia-like features. The results demonstrate that chronic hypoxia during gestation induces preeclamptic symptoms in pregnant rats via heightened ET-1 and ET-1 type A receptor-mediated signaling, providing a molecular mechanism linking gestational hypoxia and increased risk of preeclampsia.


Assuntos
Pressão Sanguínea/fisiologia , Endotelina-1/sangue , Hipóxia/metabolismo , Pré-Eclâmpsia/metabolismo , Transdução de Sinais/fisiologia , Animais , Pressão Sanguínea/efeitos dos fármacos , Modelos Animais de Doenças , Antagonistas do Receptor de Endotelina A , Feminino , Hipóxia/complicações , Hipóxia/fisiopatologia , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Rim/efeitos dos fármacos , Rim/metabolismo , Rim/fisiopatologia , Peptídeos Cíclicos/farmacologia , Placenta/metabolismo , Pré-Eclâmpsia/etiologia , Pré-Eclâmpsia/fisiopatologia , Gravidez , Proteinúria/etiologia , Proteinúria/metabolismo , Proteinúria/fisiopatologia , Ratos , Ratos Sprague-Dawley , Receptor Tipo 1 de Angiotensina/genética , Receptor Tipo 1 de Angiotensina/metabolismo , Transdução de Sinais/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...