Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 48
Filtrar
1.
Neuroendocrinology ; 114(5): 423-438, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38198758

RESUMO

INTRODUCTION: Previous studies have shown that fetal hypoxia predisposes individuals to develop addictive disorders in adulthood. However, the specific impact of maternal stress, mediated through glucocorticoids and often coexisting with fetal hypoxia, is not yet fully comprehended. METHODS: To delineate the potential effects of these pathological factors, we designed models of prenatal severe hypoxia (PSH) in conjunction with maternal stress and prenatal intrauterine ischemia (PII). We assessed the suitability of these models for our research objectives by measuring HIF1α levels and evaluating the glucocorticoid neuroendocrine system. To ascertain nicotine dependence, we employed the conditioned place aversion test and the startle response test. To identify the key factor implicated in nicotine addiction associated with PSH, we employed techniques such as Western blot, immunohistochemistry, and correlational analysis between chrna7 and nr3c1 genes across different brain structures. RESULTS: In adult rats exposed to PSH and PII, we observed increased levels of HIF1α in the hippocampus (HPC). However, the PSH group alone exhibited reduced glucocorticoid receptor levels and disturbed circadian glucocorticoid rhythms. Additionally, they displayed signs of nicotine addiction in the conditioned place aversion and startle response tests. We also observed elevated levels of phosphorylated DARPP-32 protein in the nucleus accumbens (NAc) indicated compromised glutamatergic efferent signaling. Furthermore, there was reduced expression of α7 nAChR, which modulates glutamate release, in the medial prefrontal cortex (PFC) and HPC. Correlation analysis revealed strong associations between chrna7 and nr3c1 expression in both brain structures. CONCLUSION: Perturbations in the glucocorticoid neuroendocrine system and glucocorticoid-dependent gene expression of chrna7 associated with maternal stress response to hypoxia in prenatal period favor the development of nicotine addiction in adulthood.


Assuntos
Efeitos Tardios da Exposição Pré-Natal , Estresse Psicológico , Tabagismo , Receptor Nicotínico de Acetilcolina alfa7 , Animais , Feminino , Masculino , Gravidez , Ratos , Receptor Nicotínico de Acetilcolina alfa7/genética , Receptor Nicotínico de Acetilcolina alfa7/metabolismo , Hipóxia Fetal/metabolismo , Hipóxia Fetal/complicações , Hipóxia Fetal/genética , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Efeitos Tardios da Exposição Pré-Natal/metabolismo , Ratos Sprague-Dawley , Receptores de Glucocorticoides/metabolismo , Receptores de Glucocorticoides/genética , Estresse Psicológico/metabolismo , Tabagismo/metabolismo , Tabagismo/genética , Tabagismo/complicações
2.
Am J Physiol Regul Integr Comp Physiol ; 321(6): R912-R924, 2021 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-34730023

RESUMO

We hypothesize that intrauterine hypoxia (HPX) alters the mitochondrial phenotype in fetal hearts contributing to developmental programming. Pregnant guinea pigs were exposed to normoxia (NMX) or hypoxia (HPX, 10.5% O2), starting at early [25 days (25d), 39d duration] or late gestation (50d, 14d duration). Near-term (64d) male and female fetuses were delivered by hysterotomy from anesthetized sows, and body/organ weights were measured. Left ventricles of fetal hearts were excised and frozen for measurement of expression of complex (I-V) subunits, fusion (Mfn2/OPA1) and fission (DRP1/Fis1) proteins, and enzymatic rates of I and IV from isolated mitochondrial proteins. Chronic HPX decreased fetal body weight and increased relative placenta weight regardless of timing. Early-onset HPX increased I, III, and V subunit levels, increased complex I but decreased IV activities in males but not females (all P < 0.05). Late-onset HPX decreased (P < 0.05) I, III, and V levels in both sexes but increased I and decreased IV activities in males only. Both HPX conditions decreased cardiac mitochondrial DNA content in males only. Neither early- nor late-onset HPX had any effect on Mfn2 levels but increased OPA1 in both sexes. Both HPX treatments increased DRP1/Fis1 levels in males. In females, early-onset HPX increased DRP1 with no effect on Fis1, whereas late-onset HPX increased Fis1 with no effect on DRP1. We conclude that both early- and late-onset HPX disrupts the expression/activities of select complexes that could reduce respiratory efficiency and shifts dynamics toward fission in fetal hearts. Thus, intrauterine HPX disrupts the mitochondrial phenotype predominantly in male fetal hearts, potentially altering cardiac metabolism and predisposing the offspring to heart dysfunction.


Assuntos
Complexo III da Cadeia de Transporte de Elétrons/metabolismo , Complexo II de Transporte de Elétrons/metabolismo , Complexo IV da Cadeia de Transporte de Elétrons/metabolismo , Complexo I de Transporte de Elétrons/metabolismo , Hipóxia Fetal/enzimologia , Mitocôndrias Cardíacas/enzimologia , ATPases Mitocondriais Próton-Translocadoras/metabolismo , Miócitos Cardíacos/enzimologia , Animais , Hipóxia Celular , Respiração Celular , Modelos Animais de Doenças , Dinaminas/genética , Dinaminas/metabolismo , Complexo I de Transporte de Elétrons/genética , Complexo II de Transporte de Elétrons/genética , Complexo III da Cadeia de Transporte de Elétrons/genética , Complexo IV da Cadeia de Transporte de Elétrons/genética , Feminino , Hipóxia Fetal/genética , GTP Fosfo-Hidrolases/genética , GTP Fosfo-Hidrolases/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Regulação Enzimológica da Expressão Gênica , Idade Gestacional , Cobaias , Masculino , Mitocôndrias Cardíacas/genética , Dinâmica Mitocondrial , ATPases Mitocondriais Próton-Translocadoras/genética , Fatores Sexuais
3.
Sci Rep ; 11(1): 16799, 2021 08 18.
Artigo em Inglês | MEDLINE | ID: mdl-34408219

RESUMO

Foetal hypoxia-ischaemia is a key trigger of meconium aspiration syndrome (MAS). However, many neonates develop MAS without evidence of hypoxia-ischaemia, suggesting the presence of covert but important risk variables. We evaluated the association of MAS with clinical variables, placental histopathologic findings, and inflammatory biomarkers at birth. Of 1336 symptomatic and asymptomatic term singleton neonates with meconium-stained amniotic fluid, 88 neonates (6.6%) developed MAS. Univariate analysis showed that MAS development was associated with low 1- and 5-min Apgar scores, low cord blood pH, funisitis, higher α1-acid glycoprotein levels, and higher haptoglobin levels (all p < 0.001 except for p = 0.001 for haptoglobin). Associations of MAS with caesarean delivery (p = 0.004), premature rupture of the membranes (p = 0.006), chorioamnionitis (p = 0.007), and higher C-reactive protein levels (p = 0.008) were lost when adjusted for multiple comparisons. The final multivariate model to explain MAS development comprised lower cord blood pH (odds ratio [OR] 0.58; 95% confidence interval [CI] 0.47-0.73; p < 0.001), funisitis (OR 2.45; 95% Cl 1.41-4.26; p = 0.002), and higher α1-acid glycoprotein levels (OR 1.02; 95% Cl 1.01-1.03; p = 0.001). Our data from a large cohort of neonates suggested that intrauterine inflammation is one of the key independent variables of MAS development, together with foetal hypoxia-ischaemia.


Assuntos
Hipóxia Fetal/fisiopatologia , Hipóxia-Isquemia Encefálica/fisiopatologia , Inflamação/fisiopatologia , Síndrome de Aspiração de Mecônio/fisiopatologia , Proteína C-Reativa/genética , Corioamnionite/genética , Corioamnionite/fisiopatologia , Feminino , Hipóxia Fetal/complicações , Hipóxia Fetal/genética , Humanos , Hipóxia-Isquemia Encefálica/complicações , Hipóxia-Isquemia Encefálica/genética , Recém-Nascido , Inflamação/complicações , Masculino , Síndrome de Aspiração de Mecônio/complicações , Síndrome de Aspiração de Mecônio/genética , Placenta/metabolismo , Placenta/fisiopatologia , Gravidez , Respiração Artificial , Fatores de Risco
4.
Am J Physiol Heart Circ Physiol ; 320(5): H1873-H1886, 2021 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-33739154

RESUMO

Compared with acyanotic congenital heart disease (CHD), cyanotic CHD has an increased risk of lifelong mortality and morbidity. These adverse outcomes may be attributed to delayed cardiomyocyte maturation, since the transition from a hypoxic fetal milieu to oxygen-rich postnatal environment is disrupted. We established a rodent model to replicate hypoxic myocardial conditions spanning perinatal development, and tested the hypothesis that chronic hypoxia impairs cardiac development. Pregnant mice were housed in hypoxia beginning at embryonic day 16. Pups stayed in hypoxia until postnatal day (P)8 when cardiac development is nearly complete. Global gene expression was quantified at P8 and at P30, after recovering in normoxia. Phenotypic testing included electrocardiogram, echocardiogram, and ex vivo electrophysiology study. Hypoxic P8 animals were 47% smaller than controls with preserved heart size. Gene expression was grossly altered by hypoxia at P8 (1,427 genes affected), but normalized after recovery (P30). Electrocardiograms revealed bradycardia and slowed conduction velocity in hypoxic animals at P8, with noticeable resolution after recovery (P30). Notable differences that persisted after recovery (P30) included a 65% prolongation in ventricular effective refractory period, sinus node dysfunction, 23% reduction in ejection fraction, and 16% reduction in fractional shortening in animals exposed to hypoxia. We investigated the impact of chronic hypoxia on the developing heart. Perinatal hypoxia was associated with changes in gene expression and cardiac function. Persistent changes to the electrophysiological substrate and contractile function warrant further investigation and may contribute to adverse outcomes observed in the cyanotic CHD population.NEW & NOTEWORTHY We utilized a new mouse model of chronic perinatal hypoxia to simulate the hypoxic myocardial conditions present in cyanotic congenital heart disease. Hypoxia caused numerous abnormalities in cardiomyocyte gene expression, the electrophysiologic substrate of the heart, and contractile function. Taken together, alterations observed in the neonatal period suggest delayed cardiac development immediately following hypoxia.


Assuntos
Cianose/etiologia , Coração Fetal/crescimento & desenvolvimento , Cardiopatias Congênitas/etiologia , Hipóxia/complicações , Fatores Etários , Animais , Animais Recém-Nascidos , Doença Crônica , Cianose/genética , Cianose/metabolismo , Cianose/fisiopatologia , Modelos Animais de Doenças , Feminino , Coração Fetal/metabolismo , Hipóxia Fetal/complicações , Hipóxia Fetal/genética , Hipóxia Fetal/metabolismo , Hipóxia Fetal/fisiopatologia , Regulação da Expressão Gênica no Desenvolvimento , Idade Gestacional , Cardiopatias Congênitas/genética , Cardiopatias Congênitas/metabolismo , Cardiopatias Congênitas/fisiopatologia , Frequência Cardíaca , Hipóxia/genética , Hipóxia/metabolismo , Hipóxia/fisiopatologia , Camundongos , Contração Miocárdica , Miócitos Cardíacos/metabolismo , Organogênese , Gravidez , Efeitos Tardios da Exposição Pré-Natal
5.
Mol Med Rep ; 22(5): 4173-4182, 2020 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-33000260

RESUMO

Pulmonary hypertension (PH) in newborns and adults is a disease that can lead to right heart failure and result in a shorter lifespan. PH was induced by maintaining pregnant rats in a hypoxic chamber for 4 h twice a day, from days 7­21 of pregnancy. Hypoxia was confirmed by a decrease in the partial pressure of oxygen (PaO2) and the oxygen saturation (SaO2) of arterial blood in the aorta. The body weight of newborns from hypoxic rats was ~20% decreased compared with the control newborns of normoxic rats. The vascular wall thickness/vascular diameter values of hypoxia treated pubs were increased compared with that of control newborns 7 days after birth; however, it decreased to similar levels than in the control group after 3 months, and then further decreased to significantly lower levels than in the control group at 6 months after birth. At birth, the lung tissues of newborns from hypoxic rats exhibited an increase in the levels of mRNA and proteins associated with PH such as HIF­1α, HIF­2α, V2R, TGF­ß, TNF­α, Ang­2 and α­SMA. At 3 and 6 months after birth, the levels of both V2R mRNA and protein in offspring from hypoxic rats were at least 2­fold higher, whereas the expression of all other factors decreased compared with the control offspring. By contrast, HIF­2α and Ang­2 expression levels were significantly increased in the 6­month­old control offspring from normoxic rats. V2R overexpression in pups induced by hypoxia in maternal rats was sustained until their adulthood. V2R may be a marker for detecting PH.


Assuntos
Hipóxia Fetal/complicações , Hipertensão Arterial Pulmonar/metabolismo , Receptores de Vasopressinas/genética , Receptores de Vasopressinas/metabolismo , Regulação para Cima , Angiotensina II/genética , Angiotensina II/metabolismo , Animais , Animais Recém-Nascidos , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Biomarcadores/metabolismo , Modelos Animais de Doenças , Feminino , Hipóxia Fetal/genética , Hipóxia Fetal/metabolismo , Peso Fetal , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Masculino , Hipertensão Arterial Pulmonar/etiologia , Hipertensão Arterial Pulmonar/genética , Ratos , Ratos Sprague-Dawley , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/metabolismo
6.
Genes (Basel) ; 11(3)2020 03 20.
Artigo em Inglês | MEDLINE | ID: mdl-32244901

RESUMO

Hypoxia is one of the most frequent and severe stresses to an organism's homeostatic mechanisms, and hypoxia during gestation has profound adverse effects on the heart development increasing the occurrence of congenital heart defects (CHDs). Cardiac progenitor cells (CPCs) are responsible for early heart development and the later occurrence of heart disease. However, the mechanism of how hypoxic stress affects CPC fate decisions and contributes to CHDs remains a topic of debate. Here we examined the effect of hypoxic stress on the regulations of CPC fate decisions and the potential mechanism. We found that experimental induction of hypoxic responses compromised CPC function by regulating CPC proliferation and differentiation and restraining cardiomyocyte maturation. In addition, echocardiography indicated that fetal hypoxia reduced interventricular septum thickness at diastole and the ejection time, but increased the heart rate, in mouse young adult offspring with a gender-related difference. Further study revealed that hypoxia upregulated microRNA-210 expression in Sca-1+ CPCs and impeded the cell differentiation. Blockage of microRNA-210 with LNA-anti-microRNA-210 significantly promoted differentiation of Sca-1+ CPCs into cardiomyocytes. Thus, the present findings provide clear evidence that hypoxia alters CPC fate decisions and reveal a novel mechanism of microRNA-210 in the hypoxic effect, raising the possibility of microRNA-210 as a potential therapeutic target for heart disease.


Assuntos
Diferenciação Celular , Hipóxia Fetal/metabolismo , MicroRNAs/metabolismo , Mioblastos Cardíacos/metabolismo , Miócitos Cardíacos/metabolismo , Animais , Ataxina-1/genética , Ataxina-1/metabolismo , Proliferação de Células , Células Cultivadas , Hipóxia Fetal/genética , Camundongos , MicroRNAs/genética , Mioblastos Cardíacos/citologia , Miócitos Cardíacos/citologia
7.
Pediatr Res ; 87(3): 450-455, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31185486

RESUMO

BACKGROUND: Intrauterine growth restriction (IUGR) is a pregnancy condition where fetal growth is reduced, and offspring from IUGR pregnancies are at increased risk for type II diabetes as adults. The liver is susceptible to fetal undernutrition experienced by IUGR infants and animal models of growth restriction. This study aimed to examine hepatic expression changes in a maternal nutrient restriction (MNR) mouse model of IUGR to understand fetal adaptations that influence adult metabolism. METHODS: Liver samples of male offspring from MNR (70% of ad libitum starting at E6.5) or control pregnancies were obtained at E18.5 and differential expression was assessed by RNAseq and western blots. RESULTS: Forty-nine differentially expressed (FDR < 0.1) transcripts were enriched in hypoxia-inducible pathways including Fkbp5 (1.6-fold change), Ccng2 (1.5-fold change), Pfkfb3 (1.5-fold change), Kdm3a (1.2-fold change), Btg2 (1.6-fold change), Vhl (1.3-fold change), and Hif-3a (1.3-fold change) (FDR < 0.1). Fkbp5, Pfkfb3, Kdm3a, and Hif-3a were confirmed by qPCR, but only HIF-2a (2.2-fold change, p = 0.002) and HIF-3a (1.3 p = 0.03) protein were significantly increased. CONCLUSION: Although a moderate impact, these data support evidence of fetal adaptation to reduced nutrients by increased hypoxia signaling in the liver.


Assuntos
Fenômenos Fisiológicos da Nutrição Animal , Retardo do Crescimento Fetal/metabolismo , Hipóxia Fetal/metabolismo , Fígado/metabolismo , Fenômenos Fisiológicos da Nutrição Materna , Estado Nutricional , Transdução de Sinais , Adaptação Fisiológica , Animais , Animais Recém-Nascidos , Modelos Animais de Doenças , Feminino , Retardo do Crescimento Fetal/genética , Retardo do Crescimento Fetal/fisiopatologia , Hipóxia Fetal/genética , Hipóxia Fetal/fisiopatologia , Regulação da Expressão Gênica no Desenvolvimento , Idade Gestacional , Fígado/crescimento & desenvolvimento , Masculino , Camundongos , Gravidez , Efeitos Tardios da Exposição Pré-Natal , Transdução de Sinais/genética
8.
Sci Data ; 6(1): 238, 2019 10 29.
Artigo em Inglês | MEDLINE | ID: mdl-31664036

RESUMO

The molecular mechanism of antenatal hypoxia impacting on fetal heart development and elevated risk of heart disease of adult offspring is poorly understood. We present a dataset integrating DNA methylome and transcriptome analyses of antenatal hypoxia affecting rat fetal and adult offspring hearts to understand hypoxia-mediated epigenomic reprogramming of the heart development. We showed that antenatal hypoxia not only induced DNA methylomic and transcriptomic changes in the fetal hearts, but also had a delayed and lasting effect on the adult offspring hearts. Of interest, antenatal hypoxia induced opposite changes in DNA methylation patterns in fetal and adult hearts, with a hypermethylation in the fetus and a hypomethylation in the adult. An extensive preprocessing, quality assessment, and downstream data analyses were performed on the genomic dataset so that the research community may take advantage of the public resource. These dataset could be exploited as a comprehensive resource for understanding fetal hypoxia-mediated epigenetic reprogramming in the heart development and further developmental programming of heart vulnerability to disease later in life.Figshare doi: https://doi.org/10.6084/m9.figshare.9948572.


Assuntos
Metilação de DNA , Epigenômica , Coração Fetal , Hipóxia Fetal/genética , Transcriptoma , Animais , Epigênese Genética , Coração , Ratos , Ratos Sprague-Dawley
9.
Placenta ; 83: 63-71, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-31477210

RESUMO

INTRODUCTION: The most adverse perinatal outcome of intrahepatic cholestasis of pregnancy (ICP) is sudden fetal death related to acute fetoplacental hypoxia. Corticotrophin-releasing hormone (CRH), urocortin (UCN), and Wolfram syndrome 1 (WFS1) proteins may have a compensatory response to hypoxic stress. METHODS: A total of 108 singleton pregnant women were divided into three groups: control, late-onset ICP, and early-onset ICP. Enzyme-linked immunosorbent assays were used to detected maternal serum CRH, UCN, and WFS1 levels. Western blotting and real-time polymerase chain reaction were conducted to quantify placental protein and mRNA levels of CRH, UCN, and WFS1. Pearson correlation scatterplots and Pearson correlation matrix were employed to testify the correlation. RESULTS: Placental WFS1 had a positive relation with placental UCN (r = 0.69, P < 0.05) and serum UCN (r = 0.36, P < 0.05). Placental CRH was positively correlated with maternal serum CRH (r = 0.53, P < 0.05). Maternal serum and placental levels of CRH, UCN, and WFS1 significantly increased in the early-onset ICP group compared with the control group (P < 0.05). Placental levels of UCN and WFS1 in the early-onset ICP group were significantly elevated and higher in comparison with the late-onset ICP group (P < 0.05). However, the transcriptional levels of CRH, UCN, and WFS1 were impaired in the early-onset ICP group. DISCUSSION: Our study revealed that transcription and translation of WFS1, CRH, and UCN were altered during pregnancies complicated by early-onset ICP. This disrupted compensatory response mediated by WFS1 and CRH family peptides in early-onset ICP may play a significant role in the pathogenesis of sudden fetal death in acute fetal hypoxia.


Assuntos
Colestase Intra-Hepática/metabolismo , Hormônio Liberador da Corticotropina/metabolismo , Proteínas de Membrana/metabolismo , Complicações na Gravidez/metabolismo , Adulto , Ácidos e Sais Biliares/metabolismo , Colestase Intra-Hepática/complicações , Colestase Intra-Hepática/genética , Hormônio Liberador da Corticotropina/sangue , Hormônio Liberador da Corticotropina/genética , Estresse do Retículo Endoplasmático , Feminino , Morte Fetal/etiologia , Hipóxia Fetal/etiologia , Hipóxia Fetal/genética , Hipóxia Fetal/metabolismo , Humanos , Recém-Nascido , Masculino , Proteínas de Membrana/sangue , Proteínas de Membrana/genética , Placenta/metabolismo , Gravidez , Complicações na Gravidez/genética , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Fatores de Tempo , Urocortinas/sangue , Urocortinas/genética , Urocortinas/metabolismo
10.
J Physiol ; 597(9): 2391-2401, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30791124

RESUMO

KEY POINTS: Exposure to chronic hypoxia during gestation influences long-term health and development, including reproductive capacity, across generations. If the peri-conceptual environment in the developing oviduct is affected by gestational hypoxia, then this could have implications for later fertility and the health of future generations. In the present study, we show that the oviducts of female rats exposed to chronic hypoxia in utero have reduced telomere length, decreased mitochondrial DNA biogenesis and increased oxidative stress The results of the present study show that exposure to chronic gestational hypoxia leads to accelerated ageing of the oviduct in early adulthood and they help us understand how exposure to hypoxia during development could influence reproductive health across generations. ABSTRACT: Exposure to chronic hypoxia during fetal development has important effects on immediate and long-term outcomes in offspring. Adverse impacts in adult offspring include impairment of cardiovascular function, metabolic derangement and accelerated ovarian ageing. However, it is not known whether other aspects of the female reproductive system may be similarly affected. In the present study, we examined the impact of chronic gestational hypoxia on the developing oviduct. Wistar rat dams were randomized to either normoxia (21%) or hypoxia (13%) from day 6 post-mating until delivery. Post-delivery female offspring were maintained in normoxia until 4 months of age. Oviductal gene expression was assayed at the RNA (quantitative RT-PCR) and protein (western blotting) levels. Oviductal telomere length was assayed using Southern blotting. Oviductal telomere length was reduced in the gestational hypoxia-exposed animals compared to normoxic controls (P < 0.01). This was associated with a specific post-transcriptional reduction in the KU70 subunit of DNA-pk in the gestational hypoxia-exposed group (P < 0.05). Gestational hypoxia-exposed oviducts also showed evidence of decreased mitochondrial DNA biogenesis, reduced mtDNA copy number (P < 0.05) and reduced gene expression of Tfam (P < 0.05) and Pgc1α (P < 0.05). In the hypoxia-exposed oviducts, there was upregulation of mitochondrial-specific anti-oxidant defence enzymes (MnSOD; P < 0.01). Exposure to chronic gestational hypoxia leads to accelerated ageing of the oviduct in adulthood. The oviduct plays a central role in early development as the site of gamete transport, syngamy, and early development; hence, accelerated ageing of the oviductal environment could have important implications for fertility and the health of future generations.


Assuntos
Hipóxia Fetal/fisiopatologia , Infertilidade/etiologia , Oviductos/metabolismo , Animais , DNA Mitocondrial/genética , Epigênese Genética , Feminino , Fertilidade , Hipóxia Fetal/complicações , Hipóxia Fetal/genética , Hipóxia Fetal/metabolismo , Oviductos/patologia , Estresse Oxidativo , Ratos , Ratos Wistar , Homeostase do Telômero , Transcriptoma
11.
Biol Reprod ; 100(1): 208-216, 2019 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-30085007

RESUMO

Placental hypoxia can stimulate oxidative stress and mitochondrial dysfunction reducing placental efficiency and inducing fetal growth restriction (FGR). We hypothesized that chronic hypoxia inhibits mitochondrial function in the placenta as an underlying cause of cellular mechanisms contributing to FGR. Pregnant guinea pigs were exposed to either normoxia (NMX) or hypoxia (HPX; 10.5% O2) at 25 day gestation until term (65 day). Guinea pigs were anesthetized, and fetuses and placentas were excised at either mid (40 day) or late gestation (64 day), weighed, and placental tissue stored at -80°C until assayed. Mitochondrial DNA content, protein expression of respiratory Complexes I-V, and nitration and activity rates of Complexes I and IV were measured in NMX and HPX male (N = 6 in each treatment) and female (N = 6 in each treatment) placentas. Mitochondrial density was not altered by HPX in either mid- or late-term placentas. In mid gestation, HPX slightly increased expression of Complexes I-III and V in male placentas only, but had no effect on either Complex I or IV activity rates or nitrotyrosine expression. In late gestation, HPX significantly decreased CI/CIV activity rates and increased CI/CIV nitration in male but not female placentas exhibiting a sexual dimorphism. Complex I-V expression was reduced from mid to late gestation in both male and female placentas regardless of treatment. We conclude that chronic HPX decreases mitochondrial function by inhibiting Complex I/IV activity via increased peroxynitrite in a sex-related manner. Further, there may be a progressive decrease in energy metabolism of placental cell types with gestation that increases the vulnerability of placental function to intrauterine stress.


Assuntos
Hipóxia/fisiopatologia , Mitocôndrias/fisiologia , Placenta/fisiopatologia , Efeitos Tardios da Exposição Pré-Natal , Caracteres Sexuais , Animais , DNA Mitocondrial/metabolismo , Feminino , Retardo do Crescimento Fetal/genética , Retardo do Crescimento Fetal/metabolismo , Retardo do Crescimento Fetal/patologia , Retardo do Crescimento Fetal/fisiopatologia , Hipóxia Fetal/genética , Hipóxia Fetal/metabolismo , Hipóxia Fetal/patologia , Hipóxia Fetal/fisiopatologia , Peso Fetal/fisiologia , Hipóxia/metabolismo , Masculino , Tamanho do Órgão , Estresse Oxidativo/fisiologia , Placenta/metabolismo , Placenta/patologia , Gravidez , Efeitos Tardios da Exposição Pré-Natal/metabolismo , Efeitos Tardios da Exposição Pré-Natal/patologia , Efeitos Tardios da Exposição Pré-Natal/fisiopatologia
12.
Cardiovasc Res ; 115(8): 1306-1319, 2019 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-30395198

RESUMO

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.


Assuntos
Metilação de DNA , Epigenoma , Hipóxia Fetal/genética , Cardiopatias/genética , Transcriptoma , Fatores Etários , Animais , Ilhas de CpG , Modelos Animais de Doenças , Feminino , Hipóxia Fetal/fisiopatologia , Regulação da Expressão Gênica no Desenvolvimento , Predisposição Genética para Doença , Idade Gestacional , Cardiopatias/fisiopatologia , Fenótipo , Gravidez , RNA-Seq , Ratos Sprague-Dawley , Fatores de Risco , Caracteres Sexuais , Fatores Sexuais
13.
Arkh Patol ; 80(4): 21-26, 2018.
Artigo em Russo | MEDLINE | ID: mdl-30059068

RESUMO

OBJECTIVE: To study the morphological characteristics and expression of vascular endothelial growth factor (VEGF) in the fetal testes exposed to chronic intrauterine hypoxia during pathological pregnancy in different gestation periods. MATERIAL AND METHODS: The testes from 48 male fetuses that had died in the antenatal or early neonatal period in mothers with pathological pregnancy were morphologically evaluated. RESULTS: Chronic intrauterine hypoxia was shown to be a powerful damaging factor and leads to delayed gonadal development. Histological examination of testicular tissue showed a significant reduction in the number of tubular cells per vision field, a decrease in tubular diameter and area, with the simultaneously increased area of the stroma and a larger number of vessels. Immunohistochemical study revealed the pronounced cytoplasmic expression of VEGF in testicular tissue in different gestation periods in the spermatogenic epitheliocytes, vessels, Leydig interstitial cells, while the maximal expression of this receptor was observed at 19-25 weeks' gestation, the degree of expression decreased at 26-29 weeks' gestation. CONCLUSION: Intrauterine hypoxia has a destabilizing effect on the processes of proliferation and differentiation of the spermatogenic epithelium, interstitial endocrinocytes, activates the processes of angiogenesis and the growth of connective tissue. All this can involve not only gonadal dysgenesis, but also future reproductive dysfunction. Hypoxia stimulates the expression of VEGF, whose receptors are present in almost all testicular cell populations. It can be assumed that VEGF can act as a paracrine regulator of Leydig cell activity, also as an inducer of angiogenesis, and thus play a certain role in the development of male fertility.


Assuntos
Hipóxia Fetal/fisiopatologia , Feto/fisiopatologia , Testículo/fisiopatologia , Fator A de Crescimento do Endotélio Vascular/genética , Diferenciação Celular/genética , Feminino , Hipóxia Fetal/genética , Hipóxia Fetal/mortalidade , Idade Gestacional , Humanos , Masculino , Gravidez , Testículo/crescimento & desenvolvimento
14.
J Physiol ; 596(23): 6063-6077, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-29882596

RESUMO

KEY POINTS: The cerebral response to fetal asphyxia is characterized by an upregulation of nucleic acid and chromatin modification processes, as well as a downregulation of metabolic processes at 1 h post-umbilical cord occlusion (UCO). Twenty-four hours post UCO, there was an upregulation of metabolic processes and protein modifications. UCO did not alter bacterial gene expression levels, nor did it produce a robust inflammatory response compared to maternal hypoxia. The administration of ketamine produced minimal effects on the fetal response to UCO in the cerebral cortex. ABSTRACT: Umbilical cord occlusion (UCO) is known to cause neurological disorders in the neonate. Previously, we have reported that hypoxic hypoxia (HH) stimulates the appearance of bacteria in the fetal brain and upregulates the expression of inflammatory markers in fetal cerebral cortex (CTX) and also that ketamine attenuates these responses. In the present study, we aimed to test the hypothesis that UCO, similar to HH, produces an inflammatory response in the fetal CTX and also that treatment with ketamine reduces these effects. In chronically instrumented fetal sheep (∼125 days), 30 min of partial UCO decreased fetal PaO2 levels by ∼50%. Half of the fetuses received ketamine (3 mg kg-1 ) 10 min prior to UCO (n = 4 per group). Fetal brains were collected 1 and 24 h after the experiment and mRNA was extracted and hybridized for microarray analyses. Differentially-expressed genes were analysed for significant association with gene ontologies and pathways. After 1 h, UCO upregulated nucleic acid processing and chromatin modification and downregulated metabolic processes compared to control. After 24 h, UCO upregulated metabolic and protein modification processes. Ketamine produced minimal effects. UCO did not alter the abundance of bacterial DNA in fetal brain, nor did it upregulate inflammation pathways compared to HH. We conclude that UCO produced time-dependent responses that did not include bacterial invasion or upregulation of inflammation pathways in fetal CTX. This contrasts with the response to HH, which resulted in the appearance of bacteria in the CTX and upregulated inflammation pathways. These responses in fetal CTX to oxygen deprivation are therefore modified by the maternal or placental response to the stimulus.


Assuntos
Córtex Cerebral/metabolismo , Hipóxia Fetal/genética , Feto/metabolismo , Isquemia/genética , Transcriptoma , Cordão Umbilical/irrigação sanguínea , Animais , Córtex Cerebral/efeitos dos fármacos , Córtex Cerebral/microbiologia , DNA Bacteriano , Antagonistas de Aminoácidos Excitatórios/farmacologia , Feminino , Feto/efeitos dos fármacos , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Ketamina/farmacologia , Gravidez , Ovinos , Transcriptoma/efeitos dos fármacos
15.
J Appl Oral Sci ; 26: e20170234, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29791568

RESUMO

Objectives To evaluate the association between hypoxia during embryo development and oral clefts in an animal model, and to evaluate the association between polymorphisms in the HIF-1A gene with oral clefts in human families. Material and Methods The study with the animal model used zebrafish embryos at 8 hours post-fertilization submitted to 30% and 50% hypoxia for 24 hours. At 5 days post-fertilization, the larvae were fixed. The cartilage structures were stained to evaluate craniofacial phenotypes. The family-based association study included 148 Brazilian nuclear families with oral clefts. The association between the genetic polymorphisms rs2301113 and rs2057482 in HIF-1A with oral clefts was tested. We used real time PCR genotyping approach. ANOVA with Tukey's post-test was used to compare means. The transmission/disequilibrium test was used to analyze the distortion of the inheritance of alleles from parents to their affected offspring. Results For the hypoxic animal model, the anterior portion of the ethmoid plate presented a gap in the anterior edge, forming a cleft. The hypoxia level was associated with the severity of the phenotype (p<0.0001). For the families, there was no under-transmitted allele among the affected progeny (p>0.05). Conclusion Hypoxia is involved in the oral cleft etiology, however, polymorphisms in HIF-1A are not associated with oral clefts in humans.


Assuntos
Fenda Labial/embriologia , Fenda Labial/etiologia , Fissura Palatina/embriologia , Fissura Palatina/etiologia , Hipóxia Fetal/complicações , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Polimorfismo Genético , Adolescente , Adulto , Idoso , Análise de Variância , Animais , Criança , Pré-Escolar , Modelos Animais de Doenças , Feminino , Hipóxia Fetal/genética , Estudos de Associação Genética , Humanos , Masculino , Pessoa de Meia-Idade , Reação em Cadeia da Polimerase em Tempo Real , Índice de Gravidade de Doença , Estatísticas não Paramétricas , Adulto Jovem , Peixe-Zebra
16.
J. appl. oral sci ; 26: e20170234, 2018. tab, graf
Artigo em Inglês | LILACS, BBO - Odontologia | ID: biblio-893716

RESUMO

Abstract Objectives To evaluate the association between hypoxia during embryo development and oral clefts in an animal model, and to evaluate the association between polymorphisms in the HIF-1A gene with oral clefts in human families. Material and Methods The study with the animal model used zebrafish embryos at 8 hours post-fertilization submitted to 30% and 50% hypoxia for 24 hours. At 5 days post-fertilization, the larvae were fixed. The cartilage structures were stained to evaluate craniofacial phenotypes. The family-based association study included 148 Brazilian nuclear families with oral clefts. The association between the genetic polymorphisms rs2301113 and rs2057482 in HIF-1A with oral clefts was tested. We used real time PCR genotyping approach. ANOVA with Tukey's post-test was used to compare means. The transmission/disequilibrium test was used to analyze the distortion of the inheritance of alleles from parents to their affected offspring. Results For the hypoxic animal model, the anterior portion of the ethmoid plate presented a gap in the anterior edge, forming a cleft. The hypoxia level was associated with the severity of the phenotype (p<0.0001). For the families, there was no under-transmitted allele among the affected progeny (p>0.05). Conclusion Hypoxia is involved in the oral cleft etiology, however, polymorphisms in HIF-1A are not associated with oral clefts in humans.


Assuntos
Humanos , Animais , Masculino , Feminino , Pré-Escolar , Criança , Adolescente , Adulto , Idoso , Adulto Jovem , Polimorfismo Genético , Fenda Labial/embriologia , Fenda Labial/etiologia , Fissura Palatina/embriologia , Fissura Palatina/etiologia , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Hipóxia Fetal/complicações , Índice de Gravidade de Doença , Peixe-Zebra , Análise de Variância , Estatísticas não Paramétricas , Modelos Animais de Doenças , Estudos de Associação Genética , Hipóxia Fetal/genética , Reação em Cadeia da Polimerase em Tempo Real , Pessoa de Meia-Idade
17.
Vasa ; 45(6): 439-449, 2016 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-27598052

RESUMO

Cardiovascular diseases are the main cause of mortality and morbidity in Western countries, but the underlying mechanisms are still poorly understood. Genetic polymorphisms, once thought to represent a major determinant of cardiovascular risk, individually and collectively, only explain a tiny fraction of phenotypic variation and disease risk in humans. It is now clear that non-genetic factors, i.e., factors that modify gene activity without changing the DNA sequence and that are sensitive to the environment can cause important alterations of the cardiovascular phenotype in experimental animal models and humans. Here, we will review recent studies demonstrating that distinct pathological events during the perinatal (transient perinatal hypoxemia), late foetal (preeclampsia), and early embryonic (assisted reproductive technologies) periods induce profound alterations of the cardiovascular phenotype in humans and experimental animals. Moreover, we will provide evidence that epigenetic modifications are contributing importantly to this problem and are conferring the potential for its transmission to subsequent generations.


Assuntos
Doenças Cardiovasculares/etiologia , Hipóxia Fetal/complicações , Pré-Eclâmpsia , Efeitos Tardios da Exposição Pré-Natal , Técnicas de Reprodução Assistida/efeitos adversos , Fatores Etários , Animais , Doenças Cardiovasculares/diagnóstico , Doenças Cardiovasculares/embriologia , Doenças Cardiovasculares/genética , Epigênese Genética , Feminino , Hipóxia Fetal/genética , Regulação da Expressão Gênica no Desenvolvimento , Interação Gene-Ambiente , Idade Gestacional , Humanos , Fenótipo , Pré-Eclâmpsia/genética , Gravidez , Prognóstico , Fatores de Risco
18.
Physiol Rep ; 4(6)2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-27033443

RESUMO

Transient hypoxia in pregnancy stimulates a physiological reflex response that redistributes blood flow and defends oxygen delivery to the fetal brain. We designed the present experiment to test the hypotheses that transient hypoxia produces damage of the cerebral cortex and that ketamine, an antagonist ofNMDAreceptors and a known anti-inflammatory agent, reduces the damage. Late gestation, chronically catheterized fetal sheep were subjected to a 30-min period of ventilatory hypoxia that decreased fetal PaO2from 17 ± 1 to 10 ± 1 mmHg, or normoxia (PaO217 ± 1 mmHg), with or without pretreatment (10 min before hypoxia/normoxia) with ketamine (3 mg/kg, i.v.). One day (24 h) after hypoxia/normoxia, fetal cerebral cortex was removed andmRNAextracted for transcriptomics and systems biology analysis (n = 3-5 per group). Hypoxia stimulated a transcriptomic response consistent with a reduction in cellular metabolism and an increase in inflammation. Ketamine pretreatment reduced both of these responses. The inflammation response modeled with transcriptomic systems biology was validated by immunohistochemistry and showed increased abundance of microglia/macrophages after hypoxia in the cerebral cortical tissue that ketamine significantly reduced. We conclude that transient hypoxia produces inflammation of the fetal cerebral cortex and that ketamine, in a standard clinical dose, reduces the inflammation response.


Assuntos
Anti-Inflamatórios/farmacologia , Córtex Cerebral/efeitos dos fármacos , Hipóxia Fetal/tratamento farmacológico , Hipóxia Encefálica/tratamento farmacológico , Mediadores da Inflamação/metabolismo , Ketamina/farmacologia , Fármacos Neuroprotetores/farmacologia , Animais , Córtex Cerebral/imunologia , Córtex Cerebral/metabolismo , Córtex Cerebral/patologia , Modelos Animais de Doenças , Feminino , Hipóxia Fetal/genética , Hipóxia Fetal/imunologia , Hipóxia Fetal/metabolismo , Hipóxia Fetal/patologia , Perfilação da Expressão Gênica/métodos , Regulação da Expressão Gênica , Idade Gestacional , Hipóxia Encefálica/genética , Hipóxia Encefálica/imunologia , Hipóxia Encefálica/metabolismo , Hipóxia Encefálica/patologia , Mediadores da Inflamação/imunologia , Análise de Sequência com Séries de Oligonucleotídeos , Gravidez , RNA Mensageiro/metabolismo , Ovinos , Biologia de Sistemas , Fatores de Tempo
19.
PLoS One ; 11(2): e0148465, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26859870

RESUMO

BACKGROUND: The late-gestation fetal sheep responds to hypoxia with physiological, neuroendocrine, and cellular responses that aid in fetal survival. The response of the fetus to hypoxia represents a coordinated effort to maximize oxygen transfer from the mother and minimize wasteful oxygen consumption by the fetus. While there have been many studies aimed at investigating the coordinated physiological and endocrine responses to hypoxia, and while immunohistochemical or in situ hybridization studies have revealed pathways supporting the endocrine function of the pituitary, there is little known about the coordinated cellular response of the pituitary to the hypoxia. RESULTS: Thirty min hypoxia (from 17.0±1.7 to 8.0±0.8 mm Hg, followed by 30 min normoxia) upregulated 595 and downregulated 790 genes in fetal pituitary (123-132 days' gestation; term = 147 days). Network inference of up- and down- regulated genes revealed a high degree of functional relatedness amongst the gene sets. Gene ontology analysis revealed upregulation of cellular metabolic processes (e.g., RNA synthesis, response to estrogens) and downregulation of protein phosphorylation, protein metabolism, and mitosis. Genes found to be at the center of the network of upregulated genes included genes important for purine binding and signaling. At the center of the downregulated network were genes involved in mRNA processing, DNA repair, sumoylation, and vesicular trafficking. Transcription factor analysis revealed that both up- and down-regulated gene sets are enriched for control by several transcription factors (e.g., SP1, MAZ, LEF1, NRF1, ELK1, NFAT, E12, PAX4) but not for HIF-1, which is known to be an important controller of genomic responses to hypoxia. CONCLUSIONS: The multiple analytical approaches used in this study suggests that the acute response to 30 min of transient hypoxia in the late-gestation fetus results in reduced cellular metabolism and a pattern of gene expression that is consistent with cellular oxygen and ATP starvation. In this early time point, we see a vigorous gene response. But, like the hypothalamus, the transcriptomic response is not consistent with mediation by HIF-1. If HIF-1 is a significant controller of gene expression in the fetal pituitary after hypoxia, it must be at a later time.


Assuntos
Hipóxia Fetal/genética , Hipóxia Fetal/fisiopatologia , Hipófise/fisiopatologia , Animais , Feminino , Redes Reguladoras de Genes , Idade Gestacional , Masculino , Troca Materno-Fetal/fisiologia , Modelos Genéticos , Análise de Sequência com Séries de Oligonucleotídeos , Gravidez , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase em Tempo Real , Ovinos , Transcriptoma
20.
J Pathol ; 238(5): 665-76, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26800422

RESUMO

Congenital medullary dysplasia with obstructive nephropathy is a common congenital disorder observed in paediatric patients and represents the foremost cause of renal failure. However, the molecular processes regulating normal papillary outgrowth during the postnatal period are unclear. In this study, transcriptional profiling of the renal medulla across postnatal development revealed enrichment of non-canonical Wnt signalling, vascular development, and planar cell polarity genes, all of which may contribute to perinatal medulla/papilla maturation. These pathways were investigated in a model of papillary hypoplasia with functional obstruction, the Crim1(KST264/KST264) transgenic mouse. Postnatal elongation of the renal papilla via convergent extension was unaffected in the Crim1(KST264/KST264) hypoplastic renal papilla. In contrast, these mice displayed a disorganized papillary vascular network, tissue hypoxia, and elevated Vegfa expression. In addition, we demonstrate the involvement of accompanying systemic hypoxia arising from placental insufficiency, in appropriate papillary maturation. In conclusion, this study highlights the requirement for normal vascular development in collecting duct patterning, development of appropriate nephron architecture, and perinatal papillary maturation, such that disturbances contribute to obstructive nephropathy.


Assuntos
Hipóxia Fetal/metabolismo , Medula Renal/irrigação sanguínea , Medula Renal/metabolismo , Neovascularização Patológica , Oxigênio/metabolismo , Anormalidades Urogenitais/metabolismo , Refluxo Vesicoureteral/metabolismo , Animais , Animais Recém-Nascidos , Receptores de Proteínas Morfogenéticas Ósseas/genética , Receptores de Proteínas Morfogenéticas Ósseas/metabolismo , Biologia Computacional , Modelos Animais de Doenças , Hipóxia Fetal/genética , Hipóxia Fetal/patologia , Perfilação da Expressão Gênica , Regulação da Expressão Gênica no Desenvolvimento , Genótipo , Idade Gestacional , Medula Renal/patologia , Camundongos Knockout , Neovascularização Patológica/genética , Fenótipo , Anormalidades Urogenitais/genética , Anormalidades Urogenitais/patologia , Fator A de Crescimento do Endotélio Vascular/genética , Fator A de Crescimento do Endotélio Vascular/metabolismo , Refluxo Vesicoureteral/genética , Refluxo Vesicoureteral/patologia , Via de Sinalização Wnt/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA