Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 43
Filtrar
1.
J Physiol ; 601(8): 1501-1514, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-36856073

RESUMEN

Hypoxia during pregnancy impairs uterine vascular adaptation via microRNA-210 (miR-210)-mediated mitochondrial dysfunction and mitochondrial reactive oxygen species (mtROS) generation. TET methylcytosine dioxygenase 2 (TET2) participates in regulating inflammation and oxidative stress and its deficiency contributes to the pathogenesis of multiple cardiovascular diseases. Thus, we hypothesize a role of TET2 in hypoxia/miR-210-mediated mtROS suppressing spontaneous transient outward currents (STOCs) in uterine arteries. We found that gestational hypoxia downregulated TET2 in uterine arteries of pregnant sheep and TET2 was a target of miR-210. Knockdown of TET2 with small interfering RNAs suppressed mitochondrial respiration, increased mtROS, inhibited STOCs and elevated myogenic tone. By contrast, overexpression of TET2 negated hypoxia- and miR-210-induced mtROS. The effects of TET2 knockdown in uterine arteries on mtROS, STOCs and myogenic contractions were blocked by the mitochondria-targeted antioxidant MitoQ. In addition, the recovery effects of inhibiting endogenous miR-210 with miR-210-LNA on hypoxia-induced suppression of STOCs and augmentation of myogenic tone were reversed by TET2 knockdown in uterine arteries. Together, our study reveals a novel mechanistic link between the miR-210-TET2-mtROS pathway and inhibition of STOCs and provides new insights into the understanding of uterine vascular maladaptation in pregnancy complications associated with gestational hypoxia. KEY POINTS: Gestational hypoxia downregulates TET methylcytosine dioxygenase 2 (TET2) in uterine arteries of pregnant sheep. TET2 is a downstream target of microRNA-210 (miR-210) and miR-210 mediates hypoxia-induced TET2 downregulation. Knockdown of TET2 in uterine arteries recapitulates the effect of hypoxia and miR-210 and impairs mitochondrial bioenergetics and increases mitochondrial reactive oxygen species (mtROS) . Overexpression of TET2 negates the effect of hypoxia and miR-210 on increasing mtROS. TET2 knockdown reiterates the effect of hypoxia and miR-210 and suppresses spontaneous transient outward currents (STOCs) and elevates myogenic tone, and these effects are blocked by MitoQ. Knockdown of TET2 reverses the miR-210-LNA-induced reversal of the effects of hypoxia on STOCs and myogenic tone in uterine arteries.


Asunto(s)
Dioxigenasas , MicroARNs , Embarazo , Femenino , Animales , Ovinos , Arteria Uterina/fisiología , Especies Reactivas de Oxígeno/metabolismo , Hipoxia , MicroARNs/genética , MicroARNs/metabolismo , Dioxigenasas/metabolismo , Dioxigenasas/farmacología
2.
Circulation ; 145(15): 1140-1153, 2022 04 12.
Artículo en Inglés | MEDLINE | ID: mdl-35296158

RESUMEN

BACKGROUND: Ischemic heart disease remains a leading cause of death worldwide. In this study, we test the hypothesis that microRNA-210 protects the heart from myocardial ischemia-reperfusion (IR) injury by controlling mitochondrial bioenergetics and reactive oxygen species (ROS) flux. METHODS: Myocardial infarction in an acute setting of IR was examined through comparing loss- versus gain-of-function experiments in microRNA-210-deficient and wild-type mice. Cardiac function was evaluated by echocardiography. Myocardial mitochondria bioenergetics was examined using a Seahorse XF24 Analyzer. RESULTS: MicroRNA-210 deficiency significantly exaggerated cardiac dysfunction up to 6 weeks after myocardial IR in male, but not female, mice. Intravenous injection of microRNA-210 mimic blocked the effect and recovered the increased myocardial IR injury and cardiac dysfunction. Analysis of mitochondrial metabolism revealed that microRNA-210 inhibited mitochondrial oxygen consumption, increased glycolytic activity, and reduced mitochondrial ROS flux in the heart during IR injury. Inhibition of mitochondrial ROS with MitoQ consistently reversed the effect of microRNA-210 deficiency. Mechanistically, we showed that mitochondrial glycerol-3-phosphate dehydrogenase is a novel target of microRNA-210 in the heart, and loss-of-function and gain-of-function experiments revealed that glycerol-3-phosphate dehydrogenase played a key role in the microRNA-210-mediated effect on mitochondrial metabolism and ROS flux in the setting of heart IR injury. Knockdown of glycerol-3-phosphate dehydrogenase negated microRNA-210 deficiency-induced increases in mitochondrial ROS production and myocardial infarction and improved left ventricular fractional shortening and ejection fraction after the IR treatment. CONCLUSIONS: MicroRNA-210 targeting glycerol-3-phosphate dehydrogenase controls mitochondrial bioenergetics and ROS flux and improves cardiac function in a murine model of myocardial infarction in the setting of IR injury. The findings suggest new insights into the mechanisms and therapeutic targets for treatment of ischemic heart disease.


Asunto(s)
MicroARNs , Infarto del Miocardio , Daño por Reperfusión Miocárdica , Animales , Glicerolfosfato Deshidrogenasa/metabolismo , Glicerolfosfato Deshidrogenasa/farmacología , Masculino , Ratones , MicroARNs/genética , MicroARNs/metabolismo , Mitocondrias Cardíacas/metabolismo , Infarto del Miocardio/metabolismo , Daño por Reperfusión Miocárdica/genética , Daño por Reperfusión Miocárdica/metabolismo , Daño por Reperfusión Miocárdica/prevención & control , Miocitos Cardíacos/metabolismo , Especies Reactivas de Oxígeno/metabolismo
3.
J Neuroinflammation ; 18(1): 6, 2021 Jan 05.
Artículo en Inglés | MEDLINE | ID: mdl-33402183

RESUMEN

BACKGROUND: Neonatal hypoxic-ischemic (HI) brain injury is a leading cause of acute mortality and chronic disability in newborns. Our previous studies demonstrated that HI insult significantly increased microRNA-210 (miR-210) in the brain of rat pups and inhibition of brain endogenous miR-210 by its inhibitor (LNA) provided neuroprotective effect in HI-induced brain injury. However, the molecular mechanisms underpinning this neuroprotection remain unclear. METHODS: We made a neonatal HI brain injury model in mouse pups of postnatal day 7 to uncover the mechanism of miR-210 in targeting the ten eleven translocation (TET) methylcytosine dioxygenase 2 that is a transcriptional suppressor of pro-inflammatory cytokine genes in the neonatal brain. TET2 silencing RNA was used to evaluate the role of TET2 in the neonatal HI-induced pro-inflammatory response and brain injury. MiR-210 mimic and inhibitor (LNA) were delivered into the brain of mouse pups to study the regulation of miR-210 on the expression of TET2. Luciferase reporter gene assay was performed to validate the direct binding of miR-210 to the 3' untranslated region of the TET2 transcript. Furthermore, BV2 mouse microglia cell line was employed to confirm the role of miR-210-TET2 axis in regulating pro-inflammatory response in microglia. Post-assays included chromatin immunoprecipitation (ChIP) assay, co-immunoprecipitation, RT-PCR, brain infarct assay, and neurobehavioral test. Student's t test or one-way ANOVA was used for statistical analysis. RESULTS: HI insult significantly upregulated miR-210, downregulated TET2 protein abundance, and increased NF-κB subunit p65 acetylation level and its DNA binding capacity to the interleukin 1 beta (IL-1ß) promoter in the brain of mouse pups. Inhibition of miR-210 rescued TET2 protein level from HI insult and miR-210 mimic decreased TET2 protein level in the brain of mouse pups, suggesting that TET2 is a functional target of miR-210. The co-immunoprecipitation was performed to reveal the role of TET2 in HI-induced inflammatory response in the neonatal brain. The result showed that TET2 interacted with NF-κB subunit p65 and histone deacetylase 3 (HDAC3), a co-repressor of gene transcription. Furthermore, TET2 knockdown increased transcriptional activity of acetyl-p65 on IL-1ß gene in the neonatal brain and enhanced HI-induced upregulation of acetyl-p65 level and pro-inflammatory cytokine expression. Of importance, TET2 knockdown exacerbated brain infarct size and neurological deficits and counteracted the neuroprotective effect of miR-210 inhibition. Finally, the in vitro results demonstrated that the miR-210-TET2 axis regulated pro-inflammatory response in BV2 mouse microglia cell line. CONCLUSIONS: The miR-210-TET2 axis regulates pro-inflammatory cytokine expression in microglia, contributing to neonatal HI brain injury.


Asunto(s)
Proteínas de Unión al ADN/antagonistas & inhibidores , Proteínas de Unión al ADN/biosíntesis , Regulación hacia Abajo/fisiología , Hipoxia-Isquemia Encefálica/metabolismo , Mediadores de Inflamación/metabolismo , MicroARNs/biosíntesis , Proteínas Proto-Oncogénicas/antagonistas & inhibidores , Proteínas Proto-Oncogénicas/biosíntesis , Animales , Animales Recién Nacidos , Línea Celular , Dioxigenasas , Femenino , Hipoxia-Isquemia Encefálica/patología , Masculino , Ratones
4.
J Physiol ; 596(23): 5891-5906, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-29869786

RESUMEN

KEY POINTS: Gestational hypoxia represses ten-eleven translocation methylcytosine dioxygenase 1 (TET1) expression in uterine arteries, which is recovered by inhibiting endogenous miR-210. Inhibition of miR-210 rescues BKCa channel expression and current in uterine arteries of pregnant animals acclimatized to high altitude hypoxia in a TET-dependent manner. miR-210 blockade restores BKCa channel-mediated relaxations and attenuates pressure-dependent myogenic tone in uterine arteries of pregnant animals acclimatized to high altitude. ABSTRACT: Gestational hypoxia at high altitude has profound adverse effects on the uteroplacental circulation, and is associated with increased incidence of preeclampsia and fetal intrauterine growth restriction. Previous studies demonstrated that suppression of large-conductance Ca2+ -activated K+ (BKCa ) channel function played a critical role in the maladaptation of uteroplacental circulation caused by gestational hypoxia. Yet, the mechanisms underlying gestational hypoxia-induced BKCa channel repression remain undetermined. The present study investigated a causal role of microRNA-210 (miR-210) in hypoxia-mediated repression of BKCa channel expression and function in uterine arteries using a sheep model. The results revealed that gestational hypoxia significantly decreased ten-eleven translocation methylcytosine dioxygenase 1 (TET1) expression in uterine arteries, which was recovered by inhibiting endogenous miR-210 with miR-210 locked nucleic acid (miR-210-LNA). Of importance, miR-210-LNA restored BKCa channel ß1 subunit expression in uterine arteries, which was blocked by a competitive TET inhibitor, fumarate, thus functionally linking miR-210 to the TET1-BKCa channel cascade. In addition, miR-210-LNA reversed hypoxia-mediated suppression of BKCa channel function and rescued the effect of steroid hormones in upregulating BKCa channel expression and function in uterine arteries, which were also ablated by fumarate. Collectively, the present study demonstrates a causative effect of miR-210 in the downregulation of TET1 and subsequent repression of BKCa channel expression and function, providing a novel mechanistic insight into the regulation of BKCa channel function and the molecular basis underlying the maladaptation of uterine vascular function in gestational hypoxia.


Asunto(s)
Mal de Altura/fisiopatología , MicroARNs , Canales de Potasio Calcio-Activados/fisiología , Arteria Uterina/fisiología , Animales , Femenino , Oxigenasas de Función Mixta/fisiología , Embarazo , Proteínas Proto-Oncogénicas/fisiología , Ovinos
5.
Biol Reprod ; 96(2): 424-434, 2017 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-28203702

RESUMEN

Chronic hypoxia during gestation suppresses large-conductance Ca2+-activated K+ (BKCa) channel function and impedes uterine arterial adaptation to pregnancy. This study tested the hypothesis that chronic hypoxia has a direct effect in upregulating DNA methyltransferase (DNMT) and epigenetically repressing BKCa channel beta-1 subunit (KCNMB1) expression in uterine arteries. Resistance-sized uterine arteries were isolated from near-term pregnant sheep maintained at ∼300 m above sea level or animals acclimatized to high-altitude (3,801 m) hypoxia for 110 days during gestation. For ex vivo hypoxia treatment, uterine arteries from normoxic animals were treated with 21.0% O2 or 10.5% O2 for 48 h. High-altitude hypoxia significantly upregulated DNMT3b expression and enzyme activity in uterine arteries. Similarly, ex vivo hypoxia treatment upregulated DNMT3b expression and enzyme activity that was blocked by a DNMT inhibitor 5-aza-2'-deoxycytidine (5-Aza). Of importance, 5-Aza inhibited hypoxia-induced hypermethylation of specificity protein (SP) 1 binding site at the KCNMB1 promoter and restored transcription factor binding to the KCNMB1 promoter, resulting in the recovery of KCNMB1 gene expression in uterine arteries. Furthermore, 5-Aza blocked the effect of hypoxia and rescued BKCa channel activity and reversed hypoxia-induced decrease in BKCa channel-mediated relaxations and increase in myogenic tone of uterine arteries. Collectively, these results suggest that chronic hypoxia during gestation upregulates DNMT expression and activity, resulting in hypermethylation and repression of KCNMB1 gene and BKCa channel function, impeding uterine arterial adaptation to pregnancy.


Asunto(s)
ADN (Citosina-5-)-Metiltransferasas/metabolismo , Hipoxia , Canales de Potasio Calcio-Activados/metabolismo , Arteria Uterina/metabolismo , Altitud , Animales , Azacitidina/análogos & derivados , Azacitidina/farmacología , ADN (Citosina-5-)-Metiltransferasas/genética , Decitabina , Femenino , Regulación Enzimológica de la Expresión Génica , Embarazo , Ovinos , Arteria Uterina/efectos de los fármacos , Vasodilatación/efectos de los fármacos , Vasodilatación/fisiología
6.
Int J Mol Sci ; 18(7)2017 Jun 24.
Artículo en Inglés | MEDLINE | ID: mdl-28672801

RESUMEN

Cerebral edema, primarily caused by disruption of the blood-brain barrier (BBB), is one of the serious complications associated with brain injury in neonatal hypoxic-ischemic encephalopathy (HIE). Our recent study demonstrated that the hypoxic-ischemic (HI) treatment significantly increased microRNA-210 (miR-210) in the neonatal rat brain and inhibition of miR-210 provided neuroprotection in neonatal HI brain injury. The present study aims to determine the role of miR-210 in the regulation of BBB integrity in the developing brain. miR-210 mimic was administered via intracerebroventricular injection (i.c.v.) into the brain of rat pups. Forty-eight hours after the injection, a modified Rice-Vannucci model was conducted to produce HI brain injury. Post-assays included cerebral edema analysis, western blotting, and immunofluorescence staining for serum immunoglobulin G (IgG) leakage. The results showed that miR-210 mimic exacerbated cerebral edema and IgG leakage into the brain parenchyma. In contrast, inhibition of miR-210 with its complementary locked nucleic acid oligonucleotides (miR-210-LNA) significantly reduced cerebral edema and IgG leakage. These findings suggest that miR-210 negatively regulates BBB integrity i n the neonatal brain. Mechanistically, the seed sequences of miR-210 were identified complementary to the 3' untranslated region (3' UTR) of the mRNA transcripts of tight junction protein occludin and adherens junction protein ß-catenin, indicating downstream targets of miR-210. This was further validated by in vivo data showing that miR-210 mimic significantly reduced the expression of these junction proteins in rat pup brains. Of importance, miR-210-LNA preserved the expression of junction proteins occludin and ß-catenin from neonatal HI insult. Altogether, the present study reveals a novel mechanism of miR-210 in impairing BBB integrity that contributes to cerebral edema formation after neonatal HI insult, and provides new insights in miR-210-LNA mediated neuroprotection in neonatal HI brain injury.


Asunto(s)
Barrera Hematoencefálica/metabolismo , Hipoxia-Isquemia Encefálica/genética , Hipoxia-Isquemia Encefálica/metabolismo , Uniones Intercelulares/genética , Uniones Intercelulares/metabolismo , MicroARNs/genética , Animales , Animales Recién Nacidos , Biomarcadores , Edema Encefálico/genética , Edema Encefálico/metabolismo , Edema Encefálico/patología , Modelos Animales de Enfermedad , Femenino , Regulación de la Expresión Génica , Hipoxia-Isquemia Encefálica/patología , Inmunoglobulina G/metabolismo , Masculino , Ocludina/metabolismo , Permeabilidad , Interferencia de ARN , Ratas , beta Catenina/metabolismo
7.
Neurobiol Dis ; 89: 202-12, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-26875527

RESUMEN

Perinatal hypoxic-ischemic encephalopathy (HIE) is associated with high neonatal mortality and severe long-term neurologic morbidity. Yet the mechanisms of brain injury in infants with HIE remain largely elusive. The present study determined a novel mechanism of microRNA-210 (miR-210) in silencing endogenous neuroprotection and increasing hypoxic-ischemic brain injury in neonatal rats. The study further revealed a potential therapeutic effect of miR-210 inhibition using complementary locked nucleic acid oligonucleotides (miR-210-LNA) in 10-day-old neonatal rats in the Rice-Vannucci model. The underlying mechanisms were investigated with intracerebroventricular injection (i.c.v) of miR-210 mimic, miR-210-LNA, glucocorticoid receptor (GR) agonist and antagonist. Luciferase reporter gene assay was conducted for identification of miR-210 targeting GR 3'untranslated region. The results showed that the HI treatment significantly increased miR-210 levels in the brain, and miR-210 mimic significantly decreased GR protein abundance and exacerbated HI brain injury in the pups. MiR-210-LNA administration via i.c.v. 4h after the HI insult significantly decreased brain miR-210 levels, increased GR protein abundance, reduced HI-induced neuronal death and brain infarct size, and improved long-term neurological function recovery. Of importance, the intranasal delivery of miR-210-LNA 4h after the HI insult produced similar effects in decreasing HI-induced neonatal brain injury and improving neurological function later in life. Altogether, the present study provides evidence of a novel mechanism of miR-210 in a neonatal HI brain injury model, and suggests a potential therapeutic approach of miR-210 inhibition in the treatment of neonatal HIE.


Asunto(s)
Encéfalo/metabolismo , Hipoxia-Isquemia Encefálica/metabolismo , Hipoxia-Isquemia Encefálica/prevención & control , MicroARNs/antagonistas & inhibidores , MicroARNs/metabolismo , Fármacos Neuroprotectores/administración & dosificación , Oligonucleótidos/administración & dosificación , Regiones no Traducidas 3' , Animales , Animales Recién Nacidos , Ratas , Receptores de Glucocorticoides/agonistas , Receptores de Glucocorticoides/antagonistas & inhibidores , Receptores de Glucocorticoides/genética , Receptores de Glucocorticoides/metabolismo
8.
J Pharmacol Exp Ther ; 358(2): 190-8, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27302109

RESUMEN

Dexamethasone treatment of newborn rats inhibited cardiomyocyte proliferation and stimulated premature terminal differentiation of cardiomyocytes in the developing heart. Yet mechanisms remain undetermined. The present study tested the hypothesis that the direct effect of glucocorticoid receptor-mediated epigenetic repression of cyclin D2 gene in the cardiomyocyte plays a key role in the dexamethasone-mediated effects in the developing heart. Cardiomyocytes were isolated from 2-day-old rats. Cells were stained with a cardiomyocyte marker α-actinin and a proliferation marker Ki67. Cyclin D2 expression was evaluated by Western blot and quantitative real-time polymerase chain reaction. Promoter methylation of CcnD2 was determined by methylated DNA immunoprecipitation (MeDIP). Overexpression of Cyclin D2 was conducted by transfection of FlexiCcnD2 (+CcnD2) construct. Treatment of cardiomyocytes isolated from newborn rats with dexamethasone for 48 hours significantly inhibited cardiomyocyte proliferation with increased binucleation and decreased cyclin D2 protein abundance. These effects were blocked with Ru486 (mifepristone). In addition, the dexamethasone treatment significantly increased cyclin D2 gene promoter methylation in newborn rat cardiomyocytes. 5-Aza-2'-deoxycytidine inhibited dexamethasone-mediated promoter methylation, recovered dexamethasone-induced cyclin D2 gene repression, and blocked the dexamethasone-elicited effects on cardiomyocyte proliferation and binucleation. In addition, the overexpression of cyclin D2 restored the dexamethasone-mediated inhibition of proliferation and increase in binucleation in newborn rat cardiomyocytes. The results demonstrate that dexamethasone acting on glucocorticoid receptors has a direct effect and inhibits proliferation and stimulates premature terminal differentiation of cardiomyocytes in the developing heart via epigenetic repression of cyclin D2 gene.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/genética , Ciclina D2/genética , Dexametasona/farmacología , Represión Epigenética/efectos de los fármacos , Miocitos Cardíacos/citología , Miocitos Cardíacos/efectos de los fármacos , Animales , Animales Recién Nacidos , Azacitidina/análogos & derivados , Azacitidina/farmacología , Proliferación Celular/efectos de los fármacos , Proliferación Celular/genética , Metilación de ADN/efectos de los fármacos , Metilación de ADN/genética , Decitabina , Regulación hacia Abajo/efectos de los fármacos , Regulación hacia Abajo/genética , Femenino , Miocitos Cardíacos/metabolismo , Embarazo , Regiones Promotoras Genéticas/genética , Ratas , Ratas Sprague-Dawley
9.
Biol Reprod ; 93(3): 66, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26224008

RESUMEN

Previous studies have demonstrated that perinatal nicotine exposure increased blood pressure (BP) in adult offspring. However, the underlying mechanisms were unclear. The present study tested the hypothesis that perinatal nicotine-induced programming of hypertensive response is mediated by enhanced reactive oxygen species (ROS) in the vasculature. Nicotine was administered to pregnant rats via subcutaneous osmotic mini-pumps from Day 4 of gestation to Day 10 after birth, in the absence or presence of the ROS inhibitor N-acetyl-cysteine (NAC) in the drinking water. Experiments were conducted in 8-mo-old male offspring. Perinatal nicotine treatment resulted in a significant increase in arterial ROS production in offspring, which was abrogated by NAC. Angiotensin II (Ang II)-induced BP responses were significantly higher in nicotine-treated group than in saline-treated control group, and NAC treatment blocked the nicotine-induced increase in BP response. Consistent with that, the nicotine treatment significantly increased both Ang II-induced and phorbol [12, 13]-dibutyrate (PDBu, a Prkc activator)-induced arterial contractions in adult offspring, which were blocked by NAC treatment. In addition, perinatal nicotine treatment significantly attenuated acetylcholine-induced arterial relaxation in offspring, which was also inhibited by NAC treatment. Results demonstrate that inhibition of ROS blocks the nicotine-induced increase in arterial reactivity and BP response to vasoconstrictors in adult offspring, suggesting a key role for increased oxidative stress in nicotine-induced developmental programming of hypertensive phenotype in male offspring.


Asunto(s)
Antioxidantes/farmacología , Hipertensión/inducido químicamente , Nicotina/antagonistas & inhibidores , Nicotina/toxicidad , Agonistas Nicotínicos/toxicidad , Acetilcisteína/farmacología , Angiotensina II/farmacología , Animales , Peso Corporal/efectos de los fármacos , Activación Enzimática/efectos de los fármacos , Femenino , Humanos , Relajación Muscular/efectos de los fármacos , Músculo Liso Vascular/efectos de los fármacos , Forbol 12,13-Dibutirato/farmacología , Embarazo , Efectos Tardíos de la Exposición Prenatal/prevención & control , Proteína Quinasa C/metabolismo , Ratas , Ratas Sprague-Dawley , Especies Reactivas de Oxígeno/antagonistas & inhibidores
10.
Cell Biosci ; 13(1): 58, 2023 Mar 17.
Artículo en Inglés | MEDLINE | ID: mdl-36932456

RESUMEN

BACKGROUND: Fetal hypoxia causes vital, systemic, developmental malformations in the fetus, particularly in the brain, and increases the risk of diseases in later life. We previously demonstrated that fetal hypoxia exposure increases the susceptibility of the neonatal brain to hypoxic-ischemic insult. Herein, we investigate the effect of fetal hypoxia on programming of cell-specific transcriptomes in the brain of neonatal rats. RESULTS: We obtained RNA sequencing (RNA-seq) data from neurons, microglia, oligodendrocytes, A2B5+ oligodendrocyte precursor cells, and astrocytes from male and female neonatal rats subjected either to fetal hypoxia or control conditions. Substantial transcriptomic responses to fetal hypoxia occurred in neurons, microglia, oligodendrocytes, and A2B5+ cells. Not only were the transcriptomic responses unique to each cell type, but they also occurred with a great deal of sexual dimorphism. We validated differential expression of several genes related to inflammation and cell death by Real-time Quantitative Polymerase Chain Reaction (qRT-PCR). Pathway and transcription factor motif analyses suggested that the NF-kappa B (NFκB) signaling pathway was enriched in the neonatal male brain due to fetal hypoxia, and we verified this result by transcription factor assay of NFκB-p65 in whole brain. CONCLUSIONS: Our study reveals a significant impact of fetal hypoxia on the transcriptomes of neonatal brains in a cell-specific and sex-dependent manner, and provides mechanistic insights that may help explain the development of hypoxic-ischemic sensitive phenotypes in the neonatal brain.

11.
Stroke ; 43(9): 2483-90, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22738920

RESUMEN

BACKGROUND AND PURPOSE: Maternal cigarette smoking increases the risk of neonatal morbidity. We tested the hypothesis that perinatal nicotine exposure causes heightened brain vulnerability to hypoxic-ischemic (HI) injury in neonatal rats through aberrant expression patterns of angiotensin II type 1 (AT(1)R) and type 2 (AT(2)R) receptors in the developing brain. METHODS: Nicotine was administered to pregnant rats through subcutaneous osmotic minipumps. HI brain injury was determined in 10-day-old pups. AT(1)R and AT(2)R expression patterns were assessed through Western blotting, quantitative polymerase chain reaction, immunofluorescence, and confocal imaging. RESULTS: Perinatal nicotine exposure significantly increased HI brain infarct size in male, but not female, pups. In fetal brains, nicotine caused a decrease in mRNA and protein abundance of AT(2)R but not AT(1)R. The downregulation of AT(2)R persisted in brains of male pups, and nicotine treatment resulted in a significant increase in methylation of CpG locus 3 bases upstream of TATA-box at the AT(2)R gene promoter. In female brains, there was an increase in AT(2)R but a decrease in AT(1)R expression. Both AT(1)R and AT(2)R expressed in neurons but not in astrocytes in the cortex and hippocampus. Central application of AT(1)R antagonist losartan or AT(2)R antagonist PD123319 increased HI brain infarct size in both male and female pups. In male pups, AT(2)R agonist CGP42112 abrogated nicotine-induced increase in HI brain infarction. In females, PD123319 uncovered the nicotine's effect on HI brain infarction. CONCLUSIONS: Perinatal nicotine exposure causes epigenetic repression of the AT(2)R gene in the developing brain resulting in heightened brain vulnerability to HI injury in neonatal male rats in a sex-dependent manner.


Asunto(s)
Lesión Encefálica Crónica/patología , Isquemia Encefálica/patología , Hipoxia Encefálica/patología , Nicotina/toxicidad , Agonistas Nicotínicos/toxicidad , Receptores de Angiotensina/fisiología , Angiotensina II/metabolismo , Bloqueadores del Receptor Tipo 1 de Angiotensina II/farmacología , Animales , Animales Recién Nacidos , Western Blotting , Encéfalo/patología , Metilación de ADN/efectos de los fármacos , Femenino , Retardo del Crecimiento Fetal/inducido químicamente , Retardo del Crecimiento Fetal/patología , Imidazoles/farmacología , Imidazoles/uso terapéutico , Inmunohistoquímica , Masculino , Microscopía Confocal , Embarazo , Piridinas/farmacología , Piridinas/uso terapéutico , Ratas , Ratas Sprague-Dawley , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptor de Angiotensina Tipo 1/biosíntesis , Receptor de Angiotensina Tipo 1/genética , Receptor de Angiotensina Tipo 2/biosíntesis , Receptor de Angiotensina Tipo 2/genética
12.
Br J Pharmacol ; 179(19): 4640-4654, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-35776536

RESUMEN

BACKGROUND AND PURPOSE: Hypoxia during pregnancy is associated with increased uterine vascular resistance and elevated blood pressure both in women and female sheep. A previous study demonstrated a causal role of microRNA-210 (miR-210) in gestational hypoxia-induced suppression of Ca2+ sparks/spontaneous transient outward currents (STOCs) in ovine uterine arteries, but the underlying mechanisms remain undetermined. We tested the hypothesis that miR-210 perturbs mitochondrial metabolism and increases mitochondrial reactive oxygen species (mtROS) that confer hypoxia-induced suppression of STOCs in uterine arteries. EXPERIMENTAL APPROACH: Resistance-sized uterine arteries were isolated from near-term pregnant sheep and were treated ex vivo in normoxia and hypoxia (10.5% O2 ) for 48 h. KEY RESULTS: Hypoxia increased mtROS and suppressed mitochondrial respiration in uterine arteries, which were also produced by miR-210 mimic to normoxic arteries and blocked by antagomir miR-210-LNA in hypoxic arteries. Hypoxia or miR-210 mimic inhibited Ca2+ sparks/STOCs and increased uterine arterial myogenic tone, which were inhibited by the mitochondria-targeted antioxidant MitoQ. Hypoxia and miR-210 down-regulated iron-sulfur cluster scaffold protein (ISCU) in uterine arteries and knockdown of ISCU via siRNAs suppressed mitochondrial respiration, increased mtROS, and inhibited STOCs. In addition, blockade of mitochondrial electron transport chain with antimycin and rotenone inhibited large-conductance Ca2+ -activated K+ channels, decreased STOCs and increased uterine arterial myogenic tone. CONCLUSION AND IMPLICATIONS: This study demonstrates a novel mechanistic role for the miR-210-ISCU-mtROS axis in inhibiting Ca2+ sparks/STOCs in the maladaptation of uterine arteries and provides new insights into the understanding of mitochondrial perturbations in the pathogenesis of pregnancy complications resulted from hypoxia.


Asunto(s)
MicroARNs , Arteria Uterina , Animales , Femenino , Humanos , Hipoxia/metabolismo , MicroARNs/metabolismo , Embarazo , Especies Reactivas de Oxígeno/metabolismo , Ovinos , Arteria Uterina/metabolismo
14.
Cell Mol Immunol ; 17(9): 976-991, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-31300734

RESUMEN

Neuroinflammation is a major contributor to secondary neuronal injury that accounts for a significant proportion of final brain cell loss in neonatal hypoxic-ischemic encephalopathy (HIE). However, the immunological mechanisms that underlie HIE remain unclear. MicroRNA-210 (miR-210) is the master "hypoxamir" and plays a key role in hypoxic-ischemic tissue damage. Herein, we report in an animal model of neonatal rats that HIE significantly upregulated miR-210 expression in microglia in the neonatal brain and strongly induced activated microglia. Intracerebroventricular administration of miR-210 antagomir effectively suppressed microglia-mediated neuroinflammation and significantly reduced brain injury caused by HIE. We demonstrated that miR-210 induced microglial M1 activation partly by targeting SIRT1, thereby reducing the deacetylation of the NF-κB subunit p65 and increasing NF-κB signaling activity. Thus, our study identified miR-210 as a novel regulator of microglial activation in neonatal HIE, highlighting a potential therapeutic target in the treatment of infants with hypoxic-ischemic brain injury.


Asunto(s)
Hipoxia-Isquemia Encefálica/genética , Inflamación/genética , MicroARNs/metabolismo , Microglía/metabolismo , Animales , Animales Recién Nacidos , Secuencia de Bases , Encéfalo/patología , Modelos Animales de Enfermedad , Hipoxia-Isquemia Encefálica/complicaciones , Hipoxia-Isquemia Encefálica/patología , Inflamación/complicaciones , Inflamación/patología , Activación de Macrófagos/genética , Modelos Biológicos , Neuronas/metabolismo , Oligodendroglía/metabolismo , Ratas Sprague-Dawley , Sirtuina 1/metabolismo , Regulación hacia Arriba/genética
15.
Hypertension ; 76(3): 930-942, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32683903

RESUMEN

Hypoxia during pregnancy profoundly affects uterine vascular adaptation and increases the risk of pregnancy complications, including preeclampsia and fetal intrauterine growth restriction. We recently demonstrated that increases in Ca2+ sparks and spontaneous transient outward currents (STOCs) played an essential role in pregnancy-induced uterine vascular adaptation. In the present study, we hypothesize that gestational hypoxia suppresses Ca2+ sparks/STOCs coupling leading to increased uterine vascular tone via enhanced endoplasmic reticulum (ER)/oxidative stress. Uterine arteries were obtained from nonpregnant and near-term pregnant sheep residing in low altitude or acclimatizing to high-altitude (3801 m) hypoxia for ≈110 days. High-altitude hypoxia suppressed pregnancy-induced upregulation of RyR1 and RyR2 (ryanodine receptor 1 and 2) protein abundance, Ca2+ sparks, and STOCs in uterine arteries. Inhibition of Ca2+ sparks/STOCs with the RyR inhibitor ryanodine significantly increased pressure-dependent myogenic tone in uterine arteries from low-altitude normoxic pregnant animals but not those from high-altitude hypoxic pregnant animals. Gestational hypoxia significantly increased ER/oxidative stress in uterine arteries. Of importance, the hypoxia-mediated suppression of Ca2+ sparks/STOCs and increase in myogenic tone in uterine arteries of pregnant animals were reversed by inhibiting ER/oxidative stress. Of great interest, the impaired sex hormonal regulation of STOCs in high-altitude animals was annulled by scavenging reactive oxygen species but not by inhibiting ER stress. Together, the findings reveal the differential mechanisms of ER and oxidative stresses in suppressing Ca2+ sparks/STOCs and increasing myogenic tone of uterine arteries in hypoxia during gestation, providing new insights into the understanding of pregnancy complications associated with hypoxia.


Asunto(s)
Mal de Altura/metabolismo , Señalización del Calcio/fisiología , Retículo Endoplásmico/metabolismo , Hipoxia , Canal Liberador de Calcio Receptor de Rianodina/metabolismo , Arteria Uterina/fisiología , Vasoconstricción/fisiología , Animales , Estrés del Retículo Endoplásmico , Femenino , Hipoxia/etiología , Hipoxia/metabolismo , Hipoxia/fisiopatología , Potenciales de la Membrana , Estrés Oxidativo/fisiología , Embarazo , Especies Reactivas de Oxígeno , Ovinos
16.
Front Physiol ; 10: 323, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31001129

RESUMEN

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.

17.
Mol Neurobiol ; 56(8): 5608-5625, 2019 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-30656514

RESUMEN

Neonatal hypoxic-ischemic (HI) brain injury causes significant mortality and long-term neurologic sequelae. We previously demonstrated that HI significantly increased microRNA-210 (miR-210) in the neonatal rat brain and inhibition of brain endogenous miR-210 was neuroprotective in HI brain injury. However, the molecular mechanisms underpinning this neuroprotection remain unclear. Using both in vivo and in vitro models, herein we uncover a novel mechanism mediating oxidative brain injury after neonatal HI, in which miR-210 induces mitochondrial dysfunction via downregulation of iron-sulfur cluster assembly protein (ISCU). Inhibition of miR-210 significantly ameliorates mitochondrial dysfunction, oxidative stress, and neuronal loss in the neonatal brain subjected to HI, as well as in primary cortical neurons exposed to oxygen-glucose deprivation (OGD). These effects are mediated through ISCU, in that miR-210 mimic decreases ISCU abundance in the brains of rat pups and primary cortical neurons, and inhibition of miR-210 protects ISCU against HI in vivo or OGD in vitro. Deletion of miR-210 binding sequences at the 3'UTR of ISCU transcript ablates miR-210-induced downregulation of ISCU protein abundance in PC12 cells. In primary cortical neurons, miR-210 mimic or silencing ISCU results in mitochondrial dysfunction, reactive oxygen species production, and activation of caspase-dependent death pathways. Of importance, knockdown of ISCU increases HI-induced injury in the neonatal rat brain and counteracts the neuroprotection of miR-210 inhibition. Therefore, miR-210 by downregulating ISCU and inducing mitochondrial dysfunction in neurons is a potent contributor of oxidative brain injury after neonatal HI.


Asunto(s)
Regulación hacia Abajo/genética , Hipoxia-Isquemia Encefálica/genética , Hipoxia-Isquemia Encefálica/patología , Proteínas Hierro-Azufre/genética , MicroARNs/metabolismo , Mitocondrias/metabolismo , Neuronas/patología , Animales , Animales Recién Nacidos , Lesiones Encefálicas/genética , Lesiones Encefálicas/patología , Muerte Celular , Supervivencia Celular , Células Cultivadas , Glucosa/deficiencia , Proteínas Hierro-Azufre/metabolismo , MicroARNs/genética , Neuronas/metabolismo , Estrés Oxidativo , Oxígeno , Ratas , Especies Reactivas de Oxígeno/metabolismo , Regulación hacia Arriba/genética
18.
Cells ; 8(12)2019 12 11.
Artículo en Inglés | MEDLINE | ID: mdl-31835778

RESUMEN

Antenatal hypoxia caused epigenetic reprogramming of methylome and transcriptome in the developing heart and increased the risk of heart disease later in life. Herein, we investigated the impact of gestational hypoxia in proteome and metabolome in the hearts of fetus and adult offspring. Pregnant rats were treated with normoxia or hypoxia (10.5% O2) from day 15 to 21 of gestation. Hearts were isolated from near-term fetuses and 5 month-old offspring, and proteomics and metabolomics profiling was determined. The data demonstrated that antenatal hypoxia altered proteomics and metabolomics profiling in the heart, impacting energy metabolism, lipid metabolism, oxidative stress, and inflammation-related pathways in a developmental and sex dependent manner. Of importance, integrating multi-omics data of transcriptomics, proteomics, and metabolomics profiling revealed reprogramming of the mitochondrion, especially in two clusters: (a) the cluster associated with "mitochondrial translation"/"aminoacyl t-RNA biosynthesis"/"one-carbon pool of folate"/"DNA methylation"; and (b) the cluster with "mitochondrion"/"TCA cycle and respiratory electron transfer"/"acyl-CoA dehydrogenase"/"oxidative phosphorylation"/"complex I"/"troponin myosin cardiac complex". Our study provides a powerful means of multi-omics data integration and reveals new insights into phenotypic reprogramming of the mitochondrion in the developing heart by fetal hypoxia, contributing to an increase in the heart vulnerability to disease later in life.


Asunto(s)
Corazón Fetal/crecimiento & desarrollo , Hipoxia Fetal/metabolismo , Metabolómica/métodos , Proteómica/métodos , Animales , Modelos Animales de Enfermedad , Metabolismo Energético , Femenino , Corazón Fetal/metabolismo , Metabolismo de los Lípidos , Masculino , Estrés Oxidativo , Embarazo , Ratas , Caracteres Sexuales
19.
Cardiovasc Res ; 115(8): 1306-1319, 2019 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-30395198

RESUMEN

AIMS: Antenatal hypoxia negatively impacts foetal heart development, and increases the risk of heart disease later in life. The molecular mechanisms remain largely elusive. Here, we conducted a genome-wide analysis to study the impact of antenatal hypoxia on DNA methylome and transcriptome profiling in foetal and adult offspring hearts. METHODS AND RESULTS: Pregnant rats were treated with normoxia or hypoxia (10.5% O2) from Day 15 to Day 21 of gestation. Hearts were isolated from near-term foetuses and 5-month-old male and female offsprings, and DNA methylome and RNA-seq were performed. Methylome data shows a sharp dip in CpG methylation centred at the transcription start site (TSS). CpG islands (CGIs) and CpG island shores (CGSs) within 10 kb upstream of the TSS are hypomethylated, compared with CGIs and CGSs within gene bodies. Combining transcriptome, data indicate an inverse relation between gene expression and CpG methylation around the TSS. Of interest, antenatal hypoxia induces opposite changes in methylation patterns in foetal and adult hearts, with hypermethylation in the foetus and hypomethylation in the adult. Also, there is significant sex dimorphism of changes in gene expression patterns in the adult offspring heart. Notably, pathway analysis indicates that enrichment of inflammation-related pathways are significantly greater in the adult male heart than those in the female heart. CONCLUSION: Our study provides an initial framework and new insights into foetal hypoxia-mediated epigenetic programming of pro-inflammatory phenotype in the heart development, linking antenatal stress, and developmental programming of heart vulnerability to disease later in life.


Asunto(s)
Metilación de ADN , Epigenoma , Hipoxia Fetal/genética , Cardiopatías/genética , Transcriptoma , Factores de Edad , Animales , Islas de CpG , Modelos Animales de Enfermedad , Femenino , Hipoxia Fetal/fisiopatología , Regulación del Desarrollo de la Expresión Génica , Predisposición Genética a la Enfermedad , Edad Gestacional , Cardiopatías/fisiopatología , Fenotipo , Embarazo , RNA-Seq , Ratas Sprague-Dawley , Factores de Riesgo , Caracteres Sexuales , Factores Sexuales
20.
Int J Cardiol ; 281: 82-89, 2019 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-30738609

RESUMEN

BACKGROUND: Fetal origin of adult cardiovascular disease is one of the most pressing public concerns and economic problem in modern life. Maternal cigarette smoking/nicotine abuse increases the risk of cardiovascular disease in offspring. However, the underlying mechanisms and theranostics remain unclear. We hypothesized that fetal and neonatal nicotine exposure enhances microRNA-181a (miR-181a) which targets large-conductance Ca2+-activated K+ (BKCa) channels, resulting in increased coronary vascular tone in adult offspring. METHODS: Nicotine or saline was administered to pregnant rats via subcutaneous osmotic minipumps from gestational day 4 until postnatal day 10. Experiments were conducted in adult (~6 month old) male offspring. RESULTS: Nicotine enhanced pressure-induced coronary vascular tone, which was abrogated by BKCa channel blocker. Nicotine selectively attenuated coronary BKCa ß1 but not α subunit expression. Functionally, nicotine suppressed BKCa current density and inhibited BKCa activator NS1619-induced coronary relaxations. Furthermore, activation of BKCa increased coronary flow and improved heart ischemia/reperfusion-induced infarction. Nicotine selectively enhanced miR-181a expression. MiR-181a mimic inhibited BKCa ß1 expression/channel current and decreased NS1619-induced coronary relaxation. Antioxidant eliminated the difference of BKCa current density between the saline and nicotine-treated groups and partially restored NS1619-induced relaxation in nicotine group. MiR-181a antisense decreased vascular tone and eliminated the differences between nicotine exposed and control groups. CONCLUSION: Fetal and neonatal nicotine exposure-mediated miR-181a overexpression plays an important role in nicotine-enhanced coronary vascular tone via epigenetic down-regulation of BKca channel mechanism, which provides a potentially novel therapeutic molecular target of miR-181a/BKca channels for the treatment of coronary heart ischemic disease.


Asunto(s)
Vasos Coronarios/metabolismo , Epigénesis Genética/fisiología , Subunidades alfa de los Canales de Potasio de Gran Conductancia Activados por Calcio/metabolismo , MicroARNs/biosíntesis , Nicotina/toxicidad , Vasoconstricción/fisiología , Factores de Edad , Animales , Animales Recién Nacidos , Bencimidazoles/farmacología , Vasos Coronarios/efectos de los fármacos , Regulación hacia Abajo/efectos de los fármacos , Regulación hacia Abajo/fisiología , Epigénesis Genética/efectos de los fármacos , Femenino , Subunidades alfa de los Canales de Potasio de Gran Conductancia Activados por Calcio/genética , Masculino , MicroARNs/genética , Músculo Liso Vascular/efectos de los fármacos , Músculo Liso Vascular/metabolismo , Embarazo , Ratas , Ratas Sprague-Dawley , Vasoconstricción/efectos de los fármacos , Vasodilatación/efectos de los fármacos , Vasodilatación/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA