Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
1.
J Cell Mol Med ; 24(5): 3192-3202, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-31975557

RESUMO

As a common complication of pregnancy, gestational hypoxia has been shown to predispose offspring to vascular dysfunction. Propionate, one of short-chain fatty acids, exerts cardioprotective effects via reducing blood pressure. This study examined whether prenatal hypoxia impaired propionate-stimulated large-conductance Ca2+ -activated K+ (BK) channel activities in vascular smooth muscle cells (VSMCs) of offspring. Pregnant rats were exposed to hypoxia (10.5% oxygen) and normoxia (21% oxygen) from gestational day 7-21. At 6 weeks of age, VSMCs in mesenteric arteries of offspring were analysed for BK channel functions and gene expressions. It was shown firstly that propionate could open significantly BK single channel in VSMCs in a concentration-dependent manner. Antagonists of G protein ßγ subunits and inositol trisphosphate receptor could completely suppress the activation of BK by propionate, respectively. Gαi/o and ryanodine receptor were found to participate in the stimulation on BK. Compared to the control, vasodilation and increments of BK NPo (the open probability) evoked by propionate were weakened in the offspring by prenatal hypoxia with down-regulated Gßγ and PLCß. It was indicated that prenatal hypoxia inhibited propionate-stimulated BK activities in mesenteric VSMCs of offspring via reducing expressions of Gßγ and PLCß, in which endoplasmic reticulum calcium release might be involved.


Assuntos
Hipóxia/tratamento farmacológico , Canais de Potássio Ativados por Cálcio de Condutância Alta/genética , Complicações Cardiovasculares na Gravidez/tratamento farmacológico , Efeitos Tardios da Exposição Pré-Natal/tratamento farmacológico , Propionatos/farmacologia , Animais , Pressão Sanguínea/efeitos dos fármacos , Sinalização do Cálcio/efeitos dos fármacos , Feminino , Subunidades beta da Proteína de Ligação ao GTP/genética , Humanos , Hipóxia/complicações , Hipóxia/genética , Hipóxia/patologia , Receptores de Inositol 1,4,5-Trifosfato/genética , Artérias Mesentéricas/efeitos dos fármacos , Artérias Mesentéricas/patologia , Miócitos de Músculo Liso/efeitos dos fármacos , Miócitos de Músculo Liso/patologia , Fosfolipase C beta/genética , Gravidez , Complicações Cardiovasculares na Gravidez/genética , Complicações Cardiovasculares na Gravidez/patologia , Efeitos Tardios da Exposição Pré-Natal/genética , Efeitos Tardios da Exposição Pré-Natal/metabolismo , Efeitos Tardios da Exposição Pré-Natal/patologia , Ratos
2.
Int J Med Sci ; 14(11): 1163-1172, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-29104471

RESUMO

Background: Adverse stress exposure during the early neonatal period has been shown to cause aberrant development, resulting in an increased risk of adult disease. We tested the hypothesis that neonatal exposure to lipopolysaccharide (LPS) does not alter heart function at rest condition but causes heart dysfunction under stress stimulation later in life. Methods: Saline control or LPS were administered to neonatal rats via intraperitoneal injection. Experiments were conducted in 6 week-old male and female rats. Isolated hearts were perfused in a Langendorff preparation. Results: Neonatal LPS exposure exhibited no effects on the body weight of the developing rats, but induced decreases in the left ventricle (LV) to the body weight ratio in male rats. Neonatal LPS exposure showed no effects on the baseline heart function determined by in vivo and ex vivo experiments, but caused decreases in the post-ischemic recovery of the LV function in male but not female rats. Neonatal LPS-mediated LV dysfunction was associated with an increase in myocardial infarct size and the LDH release in the male rats. Conclusion: The present study provides novel evidence that neonatal immune challenges could induce gender-dependent long-term effects on cardiac development and heart function, which reinforces the notion that adverse stress exposure during the early neonatal period can aggravate heart functions and the development of a heart ischemia-sensitive phenotype later in life.


Assuntos
Coração/fisiopatologia , Lipopolissacarídeos/toxicidade , Infarto do Miocárdio/fisiopatologia , Traumatismo por Reperfusão Miocárdica/fisiopatologia , Traumatismo por Reperfusão/fisiopatologia , Animais , Animais Recém-Nascidos , Modelos Animais de Doenças , Coração/efeitos dos fármacos , Humanos , Masculino , Infarto do Miocárdio/induzido quimicamente , Traumatismo por Reperfusão Miocárdica/induzido quimicamente , Ratos , Ratos Sprague-Dawley , Traumatismo por Reperfusão/induzido quimicamente , Caracteres Sexuais
3.
J Mol Cell Cardiol ; 91: 160-71, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26779948

RESUMO

Large studies in humans and animals have demonstrated a clear association of an adverse intrauterine environment with an increased risk of cardiovascular disease later in life. Yet mechanisms remain largely elusive. The present study tested the hypothesis that gestational hypoxia leads to promoter hypermethylation and epigenetic repression of the glucocorticoid receptor (GR) gene in the developing heart, resulting in increased heart susceptibility to ischemia and reperfusion injury in offspring. Hypoxic treatment of pregnant rats from day 15 to 21 of gestation resulted in a significant decrease of GR exon 14, 15, 16, and 17 transcripts, leading to down-regulation of GR mRNA and protein in the fetal heart. Functional cAMP-response elements (CREs) at -4408 and -3896 and Sp1 binding sites at -3425 and -3034 were identified at GR untranslated exon 1 promoters. Hypoxia significantly increased CpG methylation at the CREs and Sp1 binding sites and decreased transcription factor binding to GR exon 1 promoter, accounting for the repression of the GR gene in the developing heart. Of importance, treatment of newborn pups with 5-aza-2'-deoxycytidine reversed hypoxia-induced promoter methylation, restored GR expression and prevented hypoxia-mediated increase in ischemia and reperfusion injury of the heart in offspring. The findings demonstrate a novel mechanism of epigenetic repression of the GR gene in fetal stress-mediated programming of ischemic-sensitive phenotype in the heart.


Assuntos
Epigênese Genética , Hipóxia/genética , Traumatismo por Reperfusão Miocárdica/genética , Oxigênio/farmacologia , Receptores de Glucocorticoides/genética , Fator de Transcrição Sp1/genética , Animais , Animais Recém-Nascidos , Azacitidina/análogos & derivados , Azacitidina/farmacologia , Sítios de Ligação , Metilação de DNA/efeitos dos fármacos , Decitabina , Éxons , Feminino , Hipóxia/tratamento farmacológico , Hipóxia/metabolismo , Hipóxia/patologia , Masculino , Exposição Materna , Traumatismo por Reperfusão Miocárdica/metabolismo , Traumatismo por Reperfusão Miocárdica/patologia , Traumatismo por Reperfusão Miocárdica/prevenção & controle , Fenótipo , Gravidez , Regiões Promotoras Genéticas , Ligação Proteica , Ratos , Ratos Sprague-Dawley , Receptores de Glucocorticoides/antagonistas & inibidores , Receptores de Glucocorticoides/metabolismo , Elementos de Resposta , Fator de Transcrição Sp1/metabolismo
4.
J Reprod Dev ; 62(5): 487-493, 2016 Oct 18.
Artigo em Inglês | MEDLINE | ID: mdl-27319751

RESUMO

Although a large number of studies show that photo-period disruption potentially affects hormone secretion in mammals, information about the effects of circadian photo-period disruption during pregnancy on fetal blood reproductive hormone levels is scarce. This study used ewes and their fetuses to determine the effects of circadian photo-period disruption (deprivation of darkness) on follicle-stimulating hormone, luteinizing hormone, estradiol, and progesterone in maternal and fetal circulation at late gestation. Pregnant ewes (gestational age: 135 ± 3 days) were randomly placed into control and dark deprivation groups. The control (N = 5) and dark deprivation (N = 5) groups were exposed to a fixed 12 h light/12 h dark cycle and a 24 h constant light cycle, respectively, for 2 days. Dark deprivation up-regulated follicle-stimulating hormone and estradiol levels and down-regulated progesterone levels in both maternal and fetal circulation, and up-regulated luteinizing hormone levels in fetal but not maternal circulation. These results provide new information about how circadian photo-period disruption during pregnancy could alter the release of certain reproductive hormones into fetal blood, which may influence the development of fetal organs in utero, as well as long-term health.


Assuntos
Ritmo Circadiano , Estradiol/sangue , Hormônio Foliculoestimulante/sangue , Hormônio Luteinizante/sangue , Fotoperíodo , Progesterona/sangue , Animais , Feminino , Sangue Fetal , Regulação da Expressão Gênica no Desenvolvimento , Gravidez , Prenhez , Radioimunoensaio , Distribuição Aleatória , Ovinos , Carneiro Doméstico
5.
Cell Biochem Funct ; 33(2): 51-8, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25703688

RESUMO

Hypoxia could stimulate proliferation of mesenchymal stem cells (MSCs) under certain conditions. This study determined angiotensin II mechanisms and PI3K/AKT pathway in hypoxia-induced proliferation of MSCs. Hypoxia (3% oxygen) induced cellular proliferation in mouse MSCs and upregulated endogenous angiotensin II and angiotensin-converting enzyme in the cell culture and expression of AT1 receptors. The expressions of Sox2, not Oct4 and Rex1, were significantly increased by the hypoxia. The blockade of AT1 receptors, not AT2 receptors, depressed hypoxia induced the proliferative effects. Both hypoxia and exogenous angiotensin II activated p-AKT. Moreover, AT1 receptor inhibitor blocked the effects of hypoxia-mediated p-AKT upregulation. The data demonstrated that the hypoxia at 3% oxygen level could induce mouse MSC proliferation, probably as a result of the activation of PI3K signalling pathways via AT1 receptors.


Assuntos
Angiotensina II/metabolismo , Hipóxia Celular , Proliferação de Células , Células-Tronco Mesenquimais/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais , Animais , Células Cultivadas , Camundongos , Fatores de Transcrição/metabolismo
6.
J Endocrinol ; 244(1): 213-222, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31645018

RESUMO

Cerebral circulation is important in fetal brain development, and angiotensin II (Ang II) plays vital roles in regulation of adult cerebral circulation. However, functions of Ang II in fetal cerebral vasculature and influences of in utero hypoxia on Ang II-mediated fetal cerebral vascular responses are largely unknown. This study investigated the effects and mechanisms of in utero hypoxia on fetal middle cerebral arteries (MCA) via Ang II. Near-term ovine fetuses were exposed to in utero hypoxia, and fetal MCA responses to Ang II were tested for vascular tension, calcium transient, and molecular analysis. Ang II caused significant dose-dependent contraction in control fetal MCA. Ang II-induced MCA constriction was decreased significantly in hypoxic fetuses. Neither losartan (AT1R antagonist, 10-5 mol/L) nor PD123,319 (AT2R antagonist, 10-5 mol/L) altered Ang II-mediated contraction in fetal MCA. Phenylephrine-mediated constriction was also significantly weaker in hypoxic fetuses. Bay K8644 caused similar contractions between the two groups. Protein expression of L-type voltage-dependent calcium channels was unchanged. There were no differences in caffeine-mediated vascular tension or calcium transients. Contraction induced by PDBu (PKC agonist) was obviously weaker in hypoxic MCA. Protein expression of PKCß was reduced in the hypoxic compared with the control, along with no differences in phosphorylation levels. The results showed that fetal MCA was functionally responsive to Ang II near term. Intrauterine hypoxia reduced the vascular agonist-mediated contraction in fetal MCA, probably via decreasing PKCß and its phosphorylation, which might play protective effects on fetal cerebral circulation against transient hypoxia.


Assuntos
Angiotensina II/farmacologia , Feto/irrigação sanguínea , Hipóxia/embriologia , Proteína Quinase C beta/metabolismo , Contração Uterina/efeitos dos fármacos , Animais , Feminino , Artéria Cerebral Média/embriologia , Fosforilação/efeitos dos fármacos , Gravidez , Ovinos
7.
Mol Nutr Food Res ; 64(14): e2000196, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32506826

RESUMO

SCOPE: Although prenatal high-salt (HS) intake leads to physiological complications in the offspring, little is known regarding its effects on the offspring's glucose metabolism. Therefore, the objectives of this study are to determine the consequences of prenatal HS diet on the offspring's metabolism and to test a potential therapy. METHODS AND RESULTS: Pregnant rats are fed either a normal-salt (1% NaCl) or high-salt (8% NaCl) diet during the whole pregnancy. Experiments are conducted in five-month-old male offspring. It is found that the prenatal HS diet reduced the glucose tolerance and insulin sensitivity of the offspring. Additionally, there is down-regulation of peroxisome proliferator-activated receptor gamma coactivator 1 alpha (Ppargc1a/PPARGC1A) at the transcript and protein level, which leads to decreased mitochondrial biogenesis and oxidative respiration in skeletal muscle. Moreover, the down-regulation of Ppargc1a is accompanied by decreases in the expression of glucose transporter type 4 (Glut4). With endurance exercise training, these changes are mitigated, which ultimately resulted in improved insulin resistance. CONCLUSION: These findings suggest that prenatal HS intake induces metabolic disorders via the decreased expression of Ppargc1a in the skeletal muscle of adult offspring, providing novel information concerning the mechanisms and early prevention of metabolic diseases of fetal origins.


Assuntos
Doenças Metabólicas/etiologia , Músculo Esquelético/metabolismo , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo/metabolismo , Efeitos Tardios da Exposição Pré-Natal/etiologia , Cloreto de Sódio na Dieta/efeitos adversos , Animais , Dieta/efeitos adversos , Treino Aeróbico , Feminino , Transportador de Glucose Tipo 4/genética , Resistência à Insulina , Masculino , Doenças Metabólicas/terapia , Mitocôndrias Musculares/metabolismo , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo/genética , Condicionamento Físico Animal , Gravidez , Ratos Sprague-Dawley
8.
Reprod Toxicol ; 27(2): 149-54, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19429393

RESUMO

AIMS: Maternal cigarette smoking accompanied with fetal and neonatal growth restriction causes abnormalities in organ development in the postnatal life. The present study determined the effect of maternal administration of nicotine on the development of the kidney in rats by examining the expression of renal angiotensin II receptors at mRNA and protein levels as well as kidney weight during postnatal development. METHODS: Nicotine was administered to pregnant rats via subcutaneous osmotic minipumps throughout gestation and up to 10 days after delivery. Kidneys were removed and collected from both male and female offspring at ages of 14-day-old, 30-day-old, and 5-month-old. Maternal nicotine administration significantly reduced renal AT(2) receptor (AT(2)R) mRNA and protein abundance in both males and females at all three developmental ages examined. RESULTS: Although AT(1) receptor (AT(1)R) mRNA and protein levels were not significantly changed between the control offspring and the offspring exposed to maternal nicotine during the early developmental period, the renal AT(1)R/AT(2)R ratio was significantly increased. This was associated with a significant decrease of kidney weight in both male and female offspring. CONCLUSIONS: The results demonstrated that the development of renal angiotensin II receptor could be changed following exposure to perinatal nicotine, and such change in the kidney could be long-term in postnatal life.


Assuntos
Rim/efeitos dos fármacos , Nicotina/administração & dosagem , Agonistas Nicotínicos/administração & dosagem , Efeitos Tardios da Exposição Pré-Natal , Receptor Tipo 1 de Angiotensina/metabolismo , Receptor Tipo 2 de Angiotensina/metabolismo , Fatores Etários , Animais , Animais Recém-Nascidos , Feminino , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Idade Gestacional , Bombas de Infusão Implantáveis , Rim/embriologia , Rim/crescimento & desenvolvimento , Rim/metabolismo , Lactação , Masculino , Tamanho do Órgão/efeitos dos fármacos , Gravidez , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley , Receptor Tipo 1 de Angiotensina/genética , Receptor Tipo 2 de Angiotensina/genética
9.
Front Physiol ; 10: 323, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31001129

RESUMO

Glucocorticoid receptor (GR) signaling is critical for development and function of the heart. Our previous study demonstrated that gestational hypoxia induced epigenetic repression of the GR gene in the developing heart. The present study aims to determine that the alterations of promoter methylation level and epigenetic repression of the GR gene in the developing heart in response to maternal hypoxia is sustained in adult offspring and potential gender differences in the programming of GR gene. Pregnant rats were treated with 10.5% O2 from gestational day 15 (E15) to 21 (E21). Hearts were isolated from 5-month-old male and female offspring with the developing stage being equivalent to 18-year-old human. GR mRNA and protein abundance was determined with real time qRT-PCR and Western blot. GR gene promoter methylation and binding of transcription factors were measured with methylated DNA immunoprecipitation (MeDIP) and Chromatin immunoprecipitation (ChIP). The results showed that antenatal hypoxia significantly decreased the expression of GR mRNA and protein in the hearts of adult male offspring, but not in females, which is ascribed to the differential changes of alternative exon1 mRNA variants of GR gene in male and female hearts in response to prenatal hypoxia. In addition, the downregulation of GR expression in the male heart was correlated with increased methylation levels of CpG dinucleotides in promoters of exon 14, 15, 16, 17, and 110, which resulted in a decrease in the binding of their transcription factors. Thus, the study reveals that antenatal hypoxia results in a reprogramming and long-term change in GR gene expression in the heart by hypermethylation of GR promoter in a sex-differential pattern, which provides a novel mechanism regarding the increased vulnerability of heart later in life with exposure of prenatal hypoxia.

10.
Brain Res ; 1189: 43-50, 2008 Jan 16.
Artigo em Inglês | MEDLINE | ID: mdl-18053972

RESUMO

Prenatal exposure to nicotine can be associated with fetal abnormal development and brain damage. This study determined the effect of administration of nicotine with associated in utero hypoxia in maternal rats from early, middle, and late gestation on fetal blood hemoglobin, and expression of cholinergic receptor subtypes in the fetal brain. Our results demonstrated that maternal subcutaneous nicotine from the early gestation increased fetal hemoglobin and hematocrit, associated with reduction of PO(2). Although exposure to nicotine during late gestation had no effects on fetal brain weight, nicotine administration from the early gestation significantly decreased fetal brain muscarinic receptor (M1, M2, M3, and M4) mRNA expression, associated with restricted brain growth. Nicotine-altered muscarinic receptor subtype expression in the fetal forebrain and hindbrain showed regional differences. In addition, there were gestational differences for fetal brain muscarinic suppression by prenatal nicotine. Together, the results demonstrate that nicotine-induced in utero hypoxia is associated with poor development of muscarinic receptors in the fetal brain and restricted brain growth, and that either prolonged prenatal exposure to nicotine or critical "window" period for the brain development during pregnancy may play a role in prenatal nicotine-induced fetal muscarinic-receptor deficiency in the fetal brain.


Assuntos
Hipóxia Fetal/genética , Hipóxia Encefálica/genética , Nicotina/efeitos adversos , Efeitos Tardios da Exposição Pré-Natal/genética , RNA Mensageiro/metabolismo , Receptores Muscarínicos/genética , Animais , Encéfalo/efeitos dos fármacos , Encéfalo/embriologia , Encéfalo/fisiopatologia , Regulação para Baixo/efeitos dos fármacos , Regulação para Baixo/genética , Feminino , Hipóxia Fetal/induzido quimicamente , Hipóxia Fetal/metabolismo , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Regulação da Expressão Gênica no Desenvolvimento/genética , Hematócrito , Hipóxia Encefálica/induzido quimicamente , Hipóxia Encefálica/metabolismo , Agonistas Nicotínicos/efeitos adversos , Oxigênio/sangue , Gravidez , Efeitos Tardios da Exposição Pré-Natal/metabolismo , Efeitos Tardios da Exposição Pré-Natal/fisiopatologia , RNA Mensageiro/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Receptores Muscarínicos/deficiência , Fatores de Tempo
11.
Int J Dev Neurosci ; 26(5): 467-75, 2008 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-18407449

RESUMO

A number of studies have demonstrated the influence of nicotine on fetal development. This study determined the expression of choline acetyltransferase (ChAT), vesicular acetylcholine transporter (VAChT), and high-affinity choline transporter (CHT1) in the forebrain and hindbrain following chronic prenatal nicotine exposure in the rat fetus (maternal rats were subcutaneously injected with nicotine at different gestation periods). We also measured the effect of chronic nicotine exposure on fetal blood pO(2), pCO(2), pH, Na(+) and K(+) concentrations, as well as lactic acid levels. Maternal nicotine exposure during pregnancy was associated with a decrease in fetal pO(2) coupled with a significant increase in pCO(2) and lactic acid as well as restricted fetal growth. Additionally, maternal nicotine administration also reduced ChAT, VAChT, and CHT1 mRNA levels in the fetal brain. Nicotine-induced fetal hypoxic responses and reduced cholinergic marker expression in the brain were more severe when nicotine was started in early gestation. Our results provide new information about the effects of repeated exposure to nicotine in utero on the expression of central ChAT, VAChT, and CHT1 in the rat fetus. These results indicate that repeated hypoxic episodes or/and a direct effect of nicotine on the central cholinergic system during pregnancy may contribute to brain developmental problems in fetal origin.


Assuntos
Desenvolvimento Fetal/efeitos dos fármacos , Feto/efeitos dos fármacos , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Nicotina/toxicidade , Animais , Gasometria , Peso Corporal/efeitos dos fármacos , Encéfalo/efeitos dos fármacos , Encéfalo/embriologia , Encéfalo/metabolismo , Colina O-Acetiltransferase/genética , Eletrólitos/sangue , Feminino , Sangue Fetal/efeitos dos fármacos , Sangue Fetal/metabolismo , Hipóxia Fetal/induzido quimicamente , Feto/metabolismo , Hipóxia Encefálica/induzido quimicamente , Hipóxia Encefálica/embriologia , Exposição Materna/efeitos adversos , Proteínas de Membrana Transportadoras/genética , Nicotina/administração & dosagem , Gravidez , Prosencéfalo/efeitos dos fármacos , Prosencéfalo/embriologia , Prosencéfalo/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Rombencéfalo/efeitos dos fármacos , Rombencéfalo/embriologia , Rombencéfalo/metabolismo , Proteínas Vesiculares de Transporte de Acetilcolina/genética
12.
Neurotoxicology ; 29(4): 722-6, 2008 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-18541304

RESUMO

Previous studies have suggested that prenatal exposure to nicotine is associated with abnormal development in fetuses, including fetal brain damage. The present study determined the effect of maternal administration of nicotine during different gestational periods on brain nicotine receptor subunits in fetal rats. Subcutaneous injections of nicotine in maternal rats from the early and middle gestation decreased fetal blood PO2, increased fetal blood PCO2 and hemoglobin, and decreased fetal brain weight. The nicotinic acetylcholine receptor (nAChRs) mRNA abundance in the fetal brain was significantly changed by prenatal treatment with nicotine during pregnancy. Fetal alpha2, alpha4, alpha7, and beta2 units were significantly increased in the brain by prenatal exposure to nicotine in rat fetuses. However, the expression of mRNA of fetal brain alpha3, alpha5, beta3, and beta4 units were not changed. The results showed that prenatal nicotine can change the development of both alpha and beta subunits of nAChRs in the fetal brain at gene level in association with restriction of fetal brain growth and in utero hypoxia.


Assuntos
Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Nicotina/toxicidade , Efeitos Tardios da Exposição Pré-Natal , Receptores Nicotínicos/metabolismo , Fatores Etários , Animais , Encéfalo/embriologia , Embrião de Mamíferos , Feminino , Expressão Gênica/efeitos dos fármacos , Tamanho do Órgão/efeitos dos fármacos , Gravidez , Efeitos Tardios da Exposição Pré-Natal/induzido quimicamente , Efeitos Tardios da Exposição Pré-Natal/metabolismo , Efeitos Tardios da Exposição Pré-Natal/patologia , Subunidades Proteicas/genética , Subunidades Proteicas/metabolismo , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley , Receptores Nicotínicos/genética
13.
J Hypertens ; 36(12): 2369-2379, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30382958

RESUMO

AIMS: High-salt diet is linked to hypertension, and prenatal high-salt diet increases the risk of cardiovascular diseases in the offspring. The present study investigated whether and how prenatal high-salt diet influenced nitric oxide-mediated vasodilatation in the offspring. METHODS AND RESULTS: Pregnant rats were fed either normal-salt (1% sodium chloride) or high-salt (8% sodium chloride) diet during gestation. Experiments were conducted in 5-month-old male offspring. Sodium nitroprusside (SNP, nitric oxide donor)-induced hypotensive responses (in vivo) and vascular dilatation (in vitro) was significantly attenuated (Emax: 84 ±â€Š2 vs. 51 ±â€Š2, high-salt vs. control, P < 0.001) in the high-salt offspring, indicating reduced vascular relaxations. Pretreatment with Tempol (reactive oxygen species scavenger) alleviated this attenuation. The high-salt offspring showed an increased level of oxidative stress markers in both mesenteric arteries and plasma samples. The antioxidant activity, serum superoxide dismutase and catalase were significantly reduced, whereas malondialdehyde was increased in the high-salt offspring. O2 production, and protein expression of Nox2 and Nox4 in mesenteric arteries was significantly increased in the high-salt offspring whereas Nox1 showed no changes. The local renin-angiotensin system in mesenteric arteries was activated, associated with an increased NADPH oxidase. DNA methylation at the proximal promoter of angiotensin-converting enzyme gene in the lung was significantly increased in the high-salt offspring (P = 0.004). CONCLUSION: The results suggest that prenatal high-salt diet impairs nitric oxide-mediated vasodilatation because of the increased oxidative stress-affected renin-angiotensin system in the high-salt offspring, providing new information for understanding, and early prevention of cardiovascular diseases in fetal origins.


Assuntos
Efeitos Tardios da Exposição Pré-Natal/fisiopatologia , Sistema Renina-Angiotensina , Cloreto de Sódio na Dieta/efeitos adversos , Vasodilatação , Animais , Antioxidantes/farmacologia , Catalase/sangue , Óxidos N-Cíclicos/farmacologia , Metilação de DNA , Feminino , Hipotensão/induzido quimicamente , Hipotensão/fisiopatologia , Masculino , Malondialdeído/sangue , Artérias Mesentéricas/metabolismo , NADPH Oxidase 1/metabolismo , NADPH Oxidase 2/metabolismo , NADPH Oxidase 4/metabolismo , NADPH Oxidases/metabolismo , Óxido Nítrico/metabolismo , Doadores de Óxido Nítrico/farmacologia , Nitroprussiato/farmacologia , Estresse Oxidativo/efeitos dos fármacos , Peptidil Dipeptidase A/genética , Gravidez , Efeitos Tardios da Exposição Pré-Natal/metabolismo , Ratos , Cloreto de Sódio na Dieta/administração & dosagem , Marcadores de Spin , Superóxido Dismutase/sangue , Vasodilatação/efeitos dos fármacos
14.
Peptides ; 28(6): 1178-84, 2007 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-17513012

RESUMO

Although a number of studies have shown neural, hormonal, and behavioral capabilities in the control of body fluid regulation under conditions of dehydration in adults, limited information is available on the development of fetal functional abilities in response to osmotic challenge in rats. This study was performed to investigate the influence of maternal hypertonicity on fetal osmoregulatory capabilities at late gestational time in rats. Maternal and fetal plasma osmolality and blood sodium levels were determined and compared at continuous time points from 0.5 to 9h following maternal injection of hypertonic NaCl. Subcutaneous administration of hypertonic saline evoked a rise in plasma osmolality and sodium concentrations in maternal rats and fetuses associated with an up-regulation in angiotensinogen gene mRNA in the fetal liver and down-regulation of the same gene in the fetal brain. The increased levels of fetal blood osmolality and sodium were less than that in their mothers, and the fetus took less time to balance the enhanced osmolality and sodium concentrations. The results suggest that there may exist additional mechanisms in utero at near-term in protecting fetuses from hypertonic challenge. In addition, molecular results in the present study provide new data on fetal angiotensinogen gene expressed differently in the liver and brain under the same condition of prenatal salt loading, indicating osmotic signals of intracellular dehydration related to an acute increase in angiotensinogen mRNA in the fetal liver, and subsequent decrease in angiotensinogen mRNA levels in the fetal brain.


Assuntos
Angiotensinogênio/metabolismo , Soluções Hipertônicas/farmacologia , RNA Mensageiro/metabolismo , Angiotensinogênio/genética , Animais , Feminino , Feto/efeitos dos fármacos , Feto/metabolismo , Troca Materno-Fetal , Concentração Osmolar , Gravidez , Ratos , Ratos Sprague-Dawley , Sódio/sangue , Cloreto de Sódio/farmacologia , Fatores de Tempo
15.
Int J Clin Exp Pathol ; 10(11): 10841-10851, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-31966427

RESUMO

OBJECTIVE: To explore associations between maternal and fetal vitamin D status in preeclamptic pregnancies. METHODS: A case-control experiment was carried out with proportion ratio of 1:1 (controls: n = 60 vs cases: n = 60). Blood collection of both maternal and cord were performed before and during delivery, respectively, and 25(OH)D measurement was conducted. Difference analysis was performed according to returned data. Immunohistochemical analysis, together with semi-quantitative Western blot, was also performed to determine protein expression of vitamin D receptor in placenta and cord tissues of ESPE. RESULTS: Mean ± SD values of maternal 25(OH)D in control and PE group were 38.06 ± 6.28 and 33.05 ± 4.10, respectively, and significant differences with P < 0.0001 were found between control and PE in both continuous and categorical variables, especially in ESPE subtype (32.96 ± 4.49). The deficiency category (< 30 nmol/L) showed increased odds of PE (OR, 2.83, 95% CI, 1.32-6.08) in both maternal 25(OH)D and cord 25(OH)D in multivariable logistic regression. Semi-quantitative analysis showed that expression of placenta VDR in the ESPE subgroup was significantly higher than that in control group with P < 0.001, while expression of umbilical vein VDR in ESPE subgroup was significantly higher than that in control group with P < 0.05. CONCLUSIONS: The present study finds that lowest maternal and fetal vitamin D status in ESPE existed in the preeclampsia subsets. The VDR expression in placenta and fetus in ESPE were higher than that of normal pregnancy, which indicated that it might be related to placenta compensatory mechanism and is worthy of further research.

16.
Mol Nutr Food Res ; 61(8)2017 08.
Artigo em Inglês | MEDLINE | ID: mdl-28133948

RESUMO

SCOPE: High salt (HS) diets are related to cardiovascular diseases, and prenatal HS was suggested to increase risks of coronary artery diseases in the offspring. This study tested the hypothesis that prenatal HS may influence Adenosine-induced vasodilatation via protein kinase A (PKA) pathway in coronary arteries. METHODS AND RESULTS: Sprague-Dawley rats were fed with 8% salt diet for gestation, the control was fed with 0.3% salt diet. Coronary arteries from male adult offspring were tested for K+ channels and Adenosine signal pathways. Adenosine-mediated vasodilatation was reduced in coronary arteries in HS. There was no difference in gene expression of A2A receptors between the two groups. After pretreatment with PKA inhibitor, vasodilatation to Adenosine was decreased to a smaller extent in HS than that in control. Forskolin (activator of adenylate cyclase)-mediated vasodilatation was decreased in HS. Iberiotoxin (large-conductance Ca2+ -activated K+ channel [BK channel] inhibitor) attenuated Forskolin-induced vasodilatation in control, not in HS group. Currents of BK channels decreased in coronary artery smooth muscle cells, and PKA-modulated BK channel functions were declined. Protein levels of BK ß1 and PKA C-subunits in coronary arteries of HS offspring were reduced. CONCLUSIONS: Prenatal HS diets altered Adenosine-mediated coronary artery vasodilatation in the offspring, which was linked to downregulation of cAMP/PKA/BK channel pathway.


Assuntos
Adenosina/metabolismo , Vasos Coronários/efeitos dos fármacos , Cloreto de Sódio na Dieta/farmacologia , Vasodilatação/efeitos dos fármacos , Adenosina/farmacologia , Animais , Animais Recém-Nascidos , Vasos Coronários/metabolismo , AMP Cíclico/metabolismo , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Feminino , Canais de Potássio Ativados por Cálcio de Condutância Alta/metabolismo , Masculino , Fenômenos Fisiológicos da Nutrição Materna , Músculo Liso Vascular/citologia , Músculo Liso Vascular/efeitos dos fármacos , Músculo Liso Vascular/metabolismo , Gravidez , Efeitos Tardios da Exposição Pré-Natal , Ratos Sprague-Dawley , Receptor A2A de Adenosina/metabolismo , Transdução de Sinais
17.
Reprod Biol ; 16(3): 212-217, 2016 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-27692363

RESUMO

Melatonin is involved in the regulation of blood pressure through the receptor dependent or independent route. However, the effect of melatonin on fetal blood pressure is unknown. This study investigated the effect of melatonin on blood pressure of the late-term ovine fetus in utero. Melatonin and/or antagonists were intravenously administered into the fetuses. Mean arterial pressure and heart rate were recorded. Fetal blood samples were analyzed for biochemical parameters and hormones, including cortisol, angiotensin I, angiotensin II, aldosterone, atrial natriuretic peptide, corticotrophin-releasing hormone, adrenocorticotropic hormone, and endothelin. Fetal blood pressure was decreased following administration of melatonin, whereas it was increased following administration of luzindole, but not prazosin. Plasma level of endothelin was decreased by melatonin, which was blocked by luzindole. Our study suggested that melatonin reduced fetal blood pressure via MT1/MT2 receptors and possibly involving release of endothelin.


Assuntos
Pressão Sanguínea/efeitos dos fármacos , Melatonina/farmacologia , Receptor MT1 de Melatonina/antagonistas & inibidores , Receptor MT2 de Melatonina/antagonistas & inibidores , Transdução de Sinais/efeitos dos fármacos , Antagonistas de Receptores Adrenérgicos alfa 1/farmacologia , Hormônio Adrenocorticotrópico/sangue , Aldosterona/sangue , Angiotensina I/sangue , Angiotensina II/sangue , Animais , Fator Natriurético Atrial/sangue , Hormônio Liberador da Corticotropina/sangue , Endotelinas/sangue , Feto/efeitos dos fármacos , Frequência Cardíaca/efeitos dos fármacos , Hidrocortisona/sangue , Prazosina/farmacologia , Ovinos , Carneiro Doméstico , Triptaminas/farmacologia
18.
Atherosclerosis ; 245: 28-34, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26691907

RESUMO

BACKGROUND: Hypoxia is a critical contributor to increased risks of cardiovascular diseases, including atherosclerosis, but the detailed mechanism that hypoxia leads to atherosclerosis remains unknown. METHODS: Pregnant rats were treated with hypoxia (10.5% oxygen) during pregnancy, and HUVEC cells treated with 1% of oxygen. Blood lipids were tested at fetal stage and adult stage of offspring rats; the level of pro-inflammatory cytokines of HUVEC and offspring rats were investigated, and HIF-1α and NFκB mRNA level were also measured by Q-PCR and Elisa. RESULTS: We found that TC, LDL-C, ox-LDL-C, and the receptors of ox-LDL-C (lox-1) of the adult offspring were significantly higher than that of the control, while HDL-C was significantly reduced in hypoxia group. The internal elastic lamina was blocked by smooth muscle cells; and the migration of smooth muscle cells into the intima were observed in hypoxia offspring. Luciferase reporter gene experiment showed that HIF-1α activated NFκB transcription at four discrete binding sites of NFκBp65 promoter, although there was no obvious difference among the four discrete binding sites. Using transfection of pCDNA3.1-HIF-1α on HUVEC cells, HIF-1α significantly activated NFκB transcription at hypoxic conditions (1% O2), and concurrent with increased expression of IL-1ß and TNF-α. CONCLUSION: Hypoxia during pregnancy activated NFκB transcription to induce pro-inflammatory cytokines, leading to the early stage of atherosclerosis.


Assuntos
Aterosclerose/etiologia , Hipóxia/complicações , Lipídeos/sangue , Prenhez , Vasculite/complicações , Animais , Animais Recém-Nascidos , Aterosclerose/sangue , Aterosclerose/patologia , Células Cultivadas , Modelos Animais de Doenças , Ensaio de Imunoadsorção Enzimática , Feminino , Hipóxia/sangue , Gravidez , Ratos , Ratos Sprague-Dawley , Vasculite/sangue , Vasculite/patologia
19.
J Nutr Biochem ; 28: 121-8, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26878789

RESUMO

Adverse prenatal factors such as overtake of salt or fat food are potential risks for cardiovascular diseases in offspring. This study tested the hypothesis that prenatal high-salt (HS) diets may influence renal vascular tone and attenuates signaling pathways related to soluble guanylyl cyclase (sGC) or/and large-conductance Ca(2+)-activated K(+) (BKCa) channels in the offspring. Pregnant rats were fed either normal salt (NS) (1% NaCl) or HS (8% NaCl) diet for the whole gestation. Offspring were maintained on NS diets. Renal interlobar arteries in offspring were tested for vascular responses to phenylephrine (Phe), K(+) channels and signal pathways related to sGC. Phe induced higher vessel tension in interlobar arteries of the HS offspring. Following pretreatment with BKCa channel inhibitor iberiotoxin, Phe-mediated vasoconstrictions were decreased in HS offspring compared to NS. Phe-mediated constrictions following pretreatment with NO synthase inhibitor N(G)-nitro-l-arginine methyl ester or sGC inhibitor 1H-1,2,4-oxadiazolo-4,3-quinoxalin-1-one in the HS offspring were less sensitive than NS. The whole-cell K(+) currents and the component of BKCa channels were not changed in smooth muscle cells from interlobar arteries, whereas the K(+) currents stimulated by sGC activator BAY41-2272 were reduced in the HS offspring. The protein expressions of sGC ß1 and ß2 in the interlobar arteries of HS offspring were reduced. The results showed that chronic overintake of salt during pregnancy could increase renal vascular tone in the offspring. The affected signal pathways included down-regulation of sGC function and expression.


Assuntos
Guanilato Ciclase/metabolismo , Rim/irrigação sanguínea , Receptores Citoplasmáticos e Nucleares/metabolismo , Cloreto de Sódio na Dieta/administração & dosagem , Animais , Feminino , Gravidez , Ratos , Guanilil Ciclase Solúvel
20.
Mol Nutr Food Res ; 60(7): 1684-94, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-26991838

RESUMO

SCOPE: High-salt diet (HSD) is associated with cardiovascular diseases. This study aims at ascertaining the influence of maternal HSD on offspring's angiotensin II (ANG II)-mediated vasoconstriction and the underlying mechanisms. METHODS AND RESULTS: In comparison to a normal-salt diet, HSD used in pregnancy in rats changed the ultrastructures of the coronary artery (CA) in 5-month-old male offspring, and increased ANG II-mediated CA contractility. Measurement of [Ca(2+) ]i in CA using fluorescent fura-2, a Ca(2+) indicator, showed that ANG II-mediated increases in [Ca(2+) ]i were the same between HSD and normal-salt diet groups, but the ratio of diameter change/[Ca(2+) ]i induced by ANG II were significantly higher in HSD groups. Angiotensin II receptor type 1, not angiotensin II receptor type 2, caused ANG II-mediated vasoconstriction. Protein kinase C (PKC) inhibitor GF109203X attenuated the ANG II-mediated vasoconstriction, PKC agonist phorbol12,13-dibutyrate produced a greater contraction. There was an increase in PKCß mRNA and the corresponding protein abundance in the offspring, whereas other PKC subunits PKCα, PKCδ, and PKCε did not change. Moreover, 20 kDa myosin light chain phosphorylation levels were increased in HSD group. CONCLUSION: Maternal HSD affected the developmental programing for the offspring CA, with increased ANG II-mediated vasoconstrictions. The angiotensin II receptor type 1-PKC-20 kDa myosin light chain phosphorylation pathway was the possible mediated cellular mechanism.


Assuntos
Fenômenos Fisiológicos da Nutrição Materna , Efeitos Tardios da Exposição Pré-Natal/genética , Receptor Tipo 1 de Angiotensina/metabolismo , Cloreto de Sódio na Dieta/efeitos adversos , Vasoconstrição/efeitos dos fármacos , Angiotensina II/sangue , Animais , Aorta Torácica/efeitos dos fármacos , Aorta Torácica/metabolismo , Dieta , Feminino , Indóis/farmacologia , Masculino , Maleimidas/farmacologia , Cadeias Leves de Miosina/metabolismo , Dibutirato de 12,13-Forbol/farmacologia , Fosforilação , Gravidez , Proteína Quinase C/genética , Proteína Quinase C/metabolismo , Ratos , Ratos Sprague-Dawley , Receptor Tipo 1 de Angiotensina/genética , Renina/sangue , Sistema Renina-Angiotensina/efeitos dos fármacos , Transdução de Sinais , Sódio/sangue
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA