Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
1.
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
2.
Am J Physiol Regul Integr Comp Physiol ; 319(3): R243-R254, 2020 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-32639864

RESUMO

We hypothesized that the physiological adaptations of the fetus in response to chronic intrauterine hypoxia depend on its sex and the gestational age of exposure. Pregnant guinea pigs were exposed to room air (normoxia, NMX) or 10.5% O2 (hypoxia, HPX) at either 25 days (early onset) or 50 days (late onset) of gestation until term (~65 days). We evaluated the effects of HPX on hemodynamic and cardiac function indices using Doppler ultrasound and determined sex-related differences in near-term fetuses. Indices of uterine/umbilical artery pulsatility (PI index) and fetal heart systolic and diastolic function [Tei index and passive filling (E-wave) to filling due to atrial contraction (A-wave) (E/A ratios), respectively] were measured in utero and fetal body (FBW) and organ weights measured from extracted fetuses. Both early- and late-onset HPX decreased FBW in both males and females, had no effect on placenta weights, and increased placenta weight-to-FBW ratios. Early- but not late-onset HPX increased uterine artery PI, but neither HPX condition affected umbilical artery PI. Early-onset HPX increased left ventricle E/A ratios in both males and females, whereas late-onset HPX increased the right ventricle E/A ratio in females only. Hypoxia had no effect on the Tei index in either sex. Early- and late-onset HPX induce placental insufficiency and fetal growth restriction and increase diastolic filling depending on the sex, with female fetuses having a greater capacity than males to compensate for intrauterine hypoxia.


Assuntos
Retardo do Crescimento Fetal/fisiopatologia , Coração Fetal/fisiopatologia , Caracteres Sexuais , Artérias Umbilicais/diagnóstico por imagem , Animais , Feminino , Cobaias , Humanos , Hipóxia/fisiopatologia , Masculino , Insuficiência Placentária/fisiopatologia , Gravidez
3.
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
4.
J Physiol ; 596(23): 5535-5569, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-29633280

RESUMO

Over 30 years ago Professor David Barker first proposed the theory that events in early life could explain an individual's risk of non-communicable disease in later life: the developmental origins of health and disease (DOHaD) hypothesis. During the 1990s the validity of the DOHaD hypothesis was extensively tested in a number of human populations and the mechanisms underpinning it characterised in a range of experimental animal models. Over the past decade, researchers have sought to use this mechanistic understanding of DOHaD to develop therapeutic interventions during pregnancy and early life to improve adult health. A variety of animal models have been used to develop and evaluate interventions, each with strengths and limitations. It is becoming apparent that effective translational research requires that the animal paradigm selected mirrors the tempo of human fetal growth and development as closely as possible so that the effect of a perinatal insult and/or therapeutic intervention can be fully assessed. The guinea pig is one such animal model that over the past two decades has demonstrated itself to be a very useful platform for these important reproductive studies. This review highlights similarities in the in utero development between humans and guinea pigs, the strengths and limitations of the guinea pig as an experimental model of DOHaD and the guinea pig's potential to enhance clinical therapeutic innovation to improve human health.


Assuntos
Desenvolvimento Fetal , Modelos Animais , Pesquisa Translacional Biomédica , Animais , Feminino , Cobaias , Gravidez
5.
Am J Physiol Regul Integr Comp Physiol ; 315(6): R1232-R1241, 2018 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-30365351

RESUMO

Adverse intrauterine conditions cause fetal growth restriction and increase the risk of adult cardiovascular disease. We hypothesize that intrauterine hypoxia impairs fetal heart function, is sustained after birth, and manifests as both cardiac and mitochondrial dysfunction in offspring guinea pigs (GPs). Pregnant GPs were exposed to 10.5% O2 (HPX) at 50 days of gestation (full term = 65 days) or normoxia (NMX) for the duration of the pregnancy. Pups were allowed to deliver vaginally and raised in a NMX environment. At 90 days of age, mean arterial pressure (MAP) was measured in anesthetized GPs. NMX and prenatally HPX offspring underwent echocardiographic imaging for in vivo measurement of left ventricular cardiac morphology and function, and O2 consumption rates and complex IV enzyme activity were measured from isolated cardiomyocytes and mitochondria, respectively. Prenatal HPX increased ( P < 0.01) MAP (52.3 ± 1.3 and 58.4 ± 1.1 mmHg in NMX and HPX, respectively) and decreased ( P < 0.05) stroke volume (439.8 ± 54.5 and 289.4 ± 15.8 µl in NMX and HPX, respectively), cardiac output (94.4 ± 11.2 and 67.3 ± 3.8 ml/min in NMX and HPX, respectively), ejection fraction, and fractional shortening in male, but not female, GPs. HPX had no effect on left ventricular wall thickness or end-diastolic volume in either sex. HPX reduced mitochondrial maximal respiration and respiratory reserve capacity and complex IV activity rates in hearts of male, but not female, GPs. Prenatal HPX is a programming stimulus that increases MAP and decreases cardiac and mitochondrial function in male offspring. Sex-related differences in the contractile and mitochondrial responses suggest that female GPs are protected from cardiovascular programming of prenatal HPX.


Assuntos
Hipóxia/fisiopatologia , Mitocôndrias/metabolismo , Placenta/metabolismo , Fatores Sexuais , Animais , Modelos Animais de Doenças , Feminino , Hipóxia Fetal/metabolismo , Cobaias , Coração/fisiopatologia , Ventrículos do Coração/fisiopatologia , Gravidez , Suínos , Função Ventricular/fisiologia
6.
Am J Physiol Regul Integr Comp Physiol ; 313(4): R330-R339, 2017 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-28679680

RESUMO

Placental hypoxia is associated with maternal hypertension, placental insufficiency, and fetal growth restriction. In the pregnant guinea pig, prenatal hypoxia during early gestation inhibits cytotrophoblast invasion of spiral arteries, increases maternal blood pressure, and induces fetal growth restriction. In this study the impact of chronic maternal hypoxia on fetal heart structure was evaluated using four-dimensional echocardiography with spatiotemporal image correlation and tomographic ultrasound, and uterine and umbilical artery resistance/pulsatility indexes and fetal heart function were evaluated using pulsed-wave Doppler ultrasound. Pregnant guinea pigs were exposed to normoxia (n = 7) or hypoxia (10.5% O2, n = 9) at 28-30 days gestation, which was maintained until full term (65 days). At full term, fetal heart structure and outflow tracts were evaluated in the four-chamber view. Fetal heart diastolic function was assessed by E wave-to-A wave diastolic filling ratios (E/A ratios) of both ventricles and systolic function by the myocardial performance index (or Tie) of left ventricles of normoxic (n = 21) and hypoxic (n = 17) fetuses. There were no structural abnormalities in fetal hearts. However, hypoxia induced asymmetric fetal growth restriction and increased the placental/fetal weight compared with normoxic controls. Hypoxia increased Doppler resistance and pulsatility indexes in the uterine, but not umbilical, arteries, had no effect on the Tie index, and increased the E/A ratio in left, but not right, ventricles. Thus, prolonged hypoxia, starting at midgestation, increases uterine artery resistance and generates fetal growth restriction at full term. Furthermore, the enhanced cardiac diastolic filling with no changes in systolic function or umbilical artery resistance suggests that the fetal guinea pig systemic circulation undergoes a compensated, adaptive response to prolonged hypoxia exposure.


Assuntos
Retardo do Crescimento Fetal/fisiopatologia , Coração Fetal/fisiopatologia , Hipóxia/fisiopatologia , Complicações Cardiovasculares na Gravidez/fisiopatologia , Artéria Uterina/fisiopatologia , Útero/fisiopatologia , Animais , Ecocardiografia Tridimensional , Feminino , Retardo do Crescimento Fetal/diagnóstico por imagem , Coração Fetal/diagnóstico por imagem , Cobaias , Hipóxia/diagnóstico por imagem , Gravidez , Complicações Cardiovasculares na Gravidez/diagnóstico por imagem , Ultrassonografia Pré-Natal/métodos , Artérias Umbilicais/diagnóstico por imagem , Artérias Umbilicais/fisiopatologia , Artéria Uterina/diagnóstico por imagem , Útero/irrigação sanguínea , Útero/diagnóstico por imagem , Resistência Vascular
7.
Biol Reprod ; 95(6): 128, 2016 12.
Artigo em Inglês | MEDLINE | ID: mdl-27806942

RESUMO

Chronic placental hypoxia is one of the root causes of placental insufficiencies that result in pre-eclampsia and maternal hypertension. Chronic hypoxia causes disruption of trophoblast (TB) development, invasion into maternal decidua, and remodeling of maternal spiral arteries. The pregnant guinea pig shares several characteristics with humans such as hemomonochorial placenta, villous subplacenta, deep TB invasion, and remodeling of maternal arteries, and is an ideal animal model to study placental development. We hypothesized that chronic placental hypoxia of the pregnant guinea pig inhibits TB invasion and alters spiral artery remodeling. Time-mated pregnant guinea pigs were exposed to either normoxia (NMX) or three levels of hypoxia (HPX: 16%, 12%, or 10.5% O2) from 20 day gestation until midterm (39-40 days) or term (60-65 days). At term, HPX (10.5% O2) increased maternal arterial blood pressure (HPX 57.9 ± 2.3 vs. NMX 40.4 ± 2.3, P < 0.001), decreased fetal weight by 16.1% (P < 0.05), and increased both absolute and relative placenta weights by 10.1% and 31.8%, respectively (P < 0.05). At midterm, there was a significant increase in TB proliferation in HPX placentas as confirmed by increased PCNA and KRT7 staining and elevated ESX1 (TB marker) gene expression (P < 0.05). Additionally, quantitative image analysis revealed decreased invasion of maternal blood vessels by TB cells. In summary, this animal model of placental HPX identifies several aspects of abnormal placental development, including increased TB proliferation and decreased migration and invasion of TBs into the spiral arteries, the consequences of which are associated with maternal hypertension and fetal growth restriction.


Assuntos
Hipertensão Induzida pela Gravidez/etiologia , Hipóxia/complicações , Placenta/metabolismo , Insuficiência Placentária/etiologia , Animais , Pressão Sanguínea/fisiologia , Proliferação de Células/fisiologia , Modelos Animais de Doenças , Feminino , Cobaias , Hipertensão Induzida pela Gravidez/metabolismo , Hipertensão Induzida pela Gravidez/fisiopatologia , Hipóxia/metabolismo , Hipóxia/fisiopatologia , Queratina-7/metabolismo , Tamanho do Órgão/fisiologia , Placenta/fisiopatologia , Insuficiência Placentária/metabolismo , Insuficiência Placentária/fisiopatologia , Placentação , Gravidez , Antígeno Nuclear de Célula em Proliferação/metabolismo , Remodelação Vascular/fisiologia
8.
Reprod Sci ; 31(4): 975-986, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-37957471

RESUMO

Gestational hypoxia inhibits mitochondrial function in the fetal heart and placenta contributing to fetal growth restriction and organ dysfunction. NAD + deficiency may contribute to a metabolic deficit by inhibiting oxidative phosphorylation and ATP synthesis. We tested the effects of nicotinamide riboside (NR), an NAD + precursor, as a treatment for reversing known mitochondrial dysfunction in hypoxic fetal hearts. Pregnant guinea pigs were housed in room air (normoxia) or placed in a hypoxic chamber (10.5%O2) for the last 14 days of gestation (term = 65 days) and administered either water or NR (1.6 mg/ml) in the drinking bottle. Fetuses were excised at term, and NAD + levels of maternal liver, placenta, and fetal heart ventricles were measured. Indices of mitochondrial function (complex IV activity, sirtuin 3 activity, protein acetylation) and ATP synthesis were measured in fetal heart ventricles of NR-treated/untreated normoxic and hypoxic animals. Hypoxia reduced fetal body weight in both sexes (p = 0.01), which was prevented by NR. Hypoxia had no effect on maternal liver NAD + levels but decreased (p = 0.04) placenta NAD + levels, the latter normalized with NR treatment. Hypoxia had no effect on fetal heart NAD + but decreased (p < 0.05) mitochondrial complex IV and sirtuin 3 activities, ATP content, and increased mitochondrial acetylation, which were all normalized with maternal NR. Hypoxia increased (p < 0.05) mitochondrial acetylation in female fetal hearts but had no effect on other mitochondrial indices. We conclude that maternal NR is an effective treatment for normalizing mitochondrial dysfunction and ATP synthesis in the hypoxic fetal heart.


Assuntos
Doenças Mitocondriais , Niacinamida/análogos & derivados , Compostos de Piridínio , Sirtuína 3 , Gravidez , Masculino , Cobaias , Feminino , Animais , Humanos , NAD/metabolismo , Sirtuína 3/metabolismo , Hipóxia/metabolismo , Niacinamida/farmacologia , Mitocôndrias/metabolismo , Coração Fetal , Doenças Mitocondriais/metabolismo , Trifosfato de Adenosina/metabolismo , Hipóxia Fetal/metabolismo
9.
Reprod Sci ; 30(10): 2996-3009, 2023 10.
Artigo em Inglês | MEDLINE | ID: mdl-37138147

RESUMO

Chronic intrauterine hypoxia is a significant pregnancy complication impacting fetal heart growth, metabolism, and mitochondrial function, contributing to cardiovascular programming of the offspring. PGC1α (peroxisome proliferator-activated receptor γ co-activator 1α) is the master regulator of mitochondrial biogenesis. We investigated the effects of hypoxia on PGC1α expression following exposure at different gestational ages. Time-mated pregnant guinea pigs were exposed to normoxia (NMX, 21% O2) or hypoxia (HPX, 10.5% O2) at either 25-day (early-onset) or 50-day (late-onset) gestation, and all fetuses were extracted at term (term = ~65-day gestation). Expression of nuclear PGC1α, sirtuin 1 (SIRT1), AMP-activated protein kinase (AMPK), and mitochondrial sirtuin 3 (SIRT3) was measured, along with SIRT3 activity and mitochondrial acetylation of heart ventricles of male and female fetuses. Early-onset hypoxia increased (P<0.05) fetal cardiac nuclear PGC1α and had no effect on mitochondrial acetylation of either growth-restricted males or females. Late-onset hypoxia had either no effect or decreased (P<0.05) PCC1α expression in males and females, respectively, but increased (P<0.05) mitochondrial acetylation in both sexes. Hypoxia had variable effects on expression of SIRT1, AMPK, SIRT3, and SIRT3 activity depending on the sex. The capacity of the fetal heart to respond to hypoxia differs depending on the gestational age of exposure and sex of the fetus. Further, the effects of late-onset hypoxia on fetal heart function impose a greater risk to male than female fetuses, which has implications toward cardiovascular programming effects of the offspring.


Assuntos
Sirtuína 3 , Gravidez , Animais , Masculino , Cobaias , Feminino , Humanos , Sirtuína 3/metabolismo , Acetilação , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo/metabolismo , Proteínas Quinases Ativadas por AMP/metabolismo , Sirtuína 1/metabolismo , Hipóxia/metabolismo , Coração Fetal , Mitocôndrias/metabolismo , Hipóxia Fetal/metabolismo
10.
Pediatr Res ; 71(1): 25-31, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22289847

RESUMO

INTRODUCTION: Chronic hypoxia increases the expression of inducible nitric oxide synthase (iNOS) mRNA and protein levels in fetal guinea pig heart ventricles. Excessive generation of nitric oxide (NO) can induce nitrosative stress leading to the formation of peroxynitrite, which can upregulate the expression of matrix metalloproteinases (MMPs). This study tested the hypothesis that maternal hypoxia increases fetal cardiac MMP9 and collagen through peroxynitrite generation in fetal hearts. RESULTS: In heart ventricles, levels of malondialdehyde, 3-nitrotyrosine (3-NT), MMP9, and collagen were increased in hypoxic (HPX) vs. normoxic (NMX) fetal guinea pigs. DISCUSSION: Thus, maternal hypoxia induces oxidative-nitrosative stress and alters protein expression of the extracellular matrix (ECM) through upregulation of the iNOS pathway in fetal heart ventricles. This identifies iNOS-derived NO as an important stimulus for initiating the adverse effects of peroxynitrite in HPX fetal hearts. METHODS: Pregnant guinea pigs were exposed to normoxia (room air) or hypoxia (10.5% O(2), 14 d) before term (term ≈ 65 d) and administered water, L-N6-(1-iminoethyl)-lysine (LNIL), an iNOS inhibitor, or N-acetylcysteine (NAC), an antioxidant.


Assuntos
Colágeno/metabolismo , Coração Fetal/metabolismo , Hipóxia/metabolismo , Metaloproteinase 9 da Matriz/metabolismo , Ácido Peroxinitroso/metabolismo , Acetilcisteína/metabolismo , Animais , Peso Corporal , Feminino , Cobaias , Peroxidação de Lipídeos , Óxido Nítrico Sintase Tipo II/antagonistas & inibidores , Tamanho do Órgão , Gravidez , Tirosina/análogos & derivados , Tirosina/metabolismo
11.
Reprod Sci ; 29(10): 3015-3025, 2022 10.
Artigo em Inglês | MEDLINE | ID: mdl-35616874

RESUMO

Chronic hypoxia can cause fetal growth restriction (FGR) through placental dysfunction. Insulin-like growth factors (IGFs), such as IGF-2, play a major role in preservation of placental growth and function. We investigated the effects of chronic hypoxia and sex on protein expression of the IGF-2 pathway in placentas selected from asymmetric-FGR fetuses. Time-mated pregnant guinea pigs were assigned to normoxia (NMX, 21% O2) or hypoxia (HPX, 10.5% O2) during the last 14 days of pregnancy. Placentas were selected from male and female symmetrically grown NMX fetuses (fetal wt between 25th ile-75th ile) and HPX fetuses of asymmetric-FGR (fetal wt < 25th ile and brain:liver wt > 50th ile). Effects of HPX and sex on placenta protein expression of the IGF-2 signaling proteins (IGF-2, PI3K, AKT-P, total AKT, PCNA, a cell proliferation marker) were evaluated by immunoblotting. Effects of HPX and sex on morphometric parameters were analyzed using two-way ANOVA (p < 0.05). HPX reduced (p < 0.005) fetal wt by ~ 35% compared to NMX in both sexes. Expression of IGF-2 was lower (p = 0.029) in NMX female placentas compared to males. Despite lower NMX levels, HPX increased (p < 0.05) expression of IGF-2, AKT-P, relative AKT-P, and PCNA in female placentas only and had no effect on protein expression in male placentas. The female guinea pig placenta exhibits a greater sensitivity than males to HPX in upregulating expression of the IGF-2 axis. In addition, the sex difference in baseline IGF-2 expression suggests a greater capacity for females to increase IGF-2 in response to HPX as a placental adaptation in FGR.


Assuntos
Fator de Crescimento Insulin-Like II , Placenta , Animais , Feminino , Retardo do Crescimento Fetal/metabolismo , Feto/metabolismo , Cobaias , Humanos , Hipóxia/metabolismo , Fator de Crescimento Insulin-Like II/metabolismo , Masculino , Fosfatidilinositol 3-Quinases/metabolismo , Placenta/metabolismo , Gravidez , Antígeno Nuclear de Célula em Proliferação/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo
12.
Reprod Sci ; 29(1): 184-192, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34750769

RESUMO

Mitochondrial dysfunction is an underlying cause of childhood neurological disease secondary to the crucial role of mitochondria in proper neurodevelopment. We hypothesized that chronic intrauterine hypoxia (HPX) induces mitochondrial deficits by altering mitochondrial biogenesis and dynamics in the fetal brain. Pregnant guinea pigs were exposed to either normoxia (NMX, 21%O2) or HPX (10.5%O2) starting at 28-day (early onset, EO-HPX) or 50-day (late onset, LO-HPX) gestation until term (65 days). Near-term male and female fetuses were extracted from anesthetized sows, and mitochondria were isolated from excised fetal forebrains (n = 6/group). Expression of mitochondrial complex subunits I-V (CI-CV), fission (Drp-1), and fusion (Mfn-2) proteins was measured by Western blot. CI and CIV enzyme activities were measured by colorimetric assays. Chronic HPX reduced fetal body wts and increased (P < 0.05) brain/body wt ratios of both sexes. CV subunit levels were increased in EO-HPX males only and CII levels increased in LO-HPX females only compared to NMX. Both EO- and LO-HPX decreased CIV activity in both sexes but had no effect on CI activity. EO-HPX increased Drp1 and decreased Mfn2 levels in males, while LO-HPX had no effect on either protein levels. In females, both EO-HPX and LO-HPX increased Drp1 but had no effect on Mfn2 levels. Chronic HPX alters abundance and activity of select complex subunits and shifts mitochondrial dynamics toward fission in a sex-dependent manner in the fetal guinea pig brain. This may be an underlying mechanism of reduced respiratory efficiency leading to disrupted metabolism and increased vulnerability to a second neurological injury at the time of birth in HPX fetal brains.


Assuntos
Complexo IV da Cadeia de Transporte de Elétrons/metabolismo , Hipóxia Fetal/metabolismo , Prosencéfalo/metabolismo , Animais , Complexo I de Transporte de Elétrons/metabolismo , Feminino , Cobaias , Dinâmica Mitocondrial , Gravidez
13.
Oxid Med Cell Longev ; 2019: 7210249, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31249648

RESUMO

Chronic intrauterine hypoxia is a programming stimulus of cardiovascular dysfunction. While the fetal heart adapts to the reduced oxygenation, the offspring heart becomes vulnerable to subsequent metabolic challenges as an adult. Cardiac mitochondria are key organelles responsible for an efficient energy supply but are subject to damage under hypoxic conditions. We propose that intrauterine hypoxia alters mitochondrial function as an underlying programming mechanism of contractile dysfunction in the offspring. Indices of mitochondrial function such as mitochondrial DNA content, Complex (C) I-V expression, and CI/CIV enzyme activity were measured in hearts of male and female offspring at 90 days old exposed to prenatal hypoxia (10.5% O2) for 14 d prior to term (65 d). Both left ventricular tissue and cardiomyocytes exhibited decreased mitochondrial DNA content, expression of CIV, and CI/CIV activity in male hearts. In female cardiomyocytes, hypoxia had no effect on protein expression of CI-CV nor on CI/CIV activity. This study suggests that chronic intrauterine hypoxia alters the intrinsic properties of select respiratory complexes as a programming mechanism of cardiac dysfunction in the offspring. Sex differences in mitochondrial function may underlie the increased vulnerability of age-matched males compared to females in cardiovascular disease and heart failure.


Assuntos
Desenvolvimento Fetal , Coração Fetal/patologia , Hipóxia Fetal/fisiopatologia , Cardiopatias/patologia , Mitocôndrias Cardíacas/patologia , Proteínas Mitocondriais/metabolismo , Efeitos Tardios da Exposição Pré-Natal/epidemiologia , Caracteres Sexuais , Animais , Animais Recém-Nascidos , Feminino , Coração Fetal/metabolismo , Cobaias , Cardiopatias/metabolismo , Masculino , Mitocôndrias Cardíacas/metabolismo , Gravidez , Efeitos Tardios da Exposição Pré-Natal/patologia
14.
Am J Obstet Gynecol ; 199(1): 78.e1-6, 2008 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-18279828

RESUMO

OBJECTIVE: Intrauterine infection increases proinflammatory cytokines in the fetus. We hypothesize that proinflammatory cytokines and matrix metalloproteinases (MMPs) are upregulated in fetal hearts in response to hypoxic stress. STUDY DESIGN: Timed-pregnant guinea pigs were exposed to either hypoxia (10.5% O(2), 14 day) or normoxia (room air). Left ventricles of fetal hearts were excised from anesthetized age-matched fetuses and frozen until ready for study. Messenger RNA of pro- (TNF-alpha, IL-6, IL-1beta) and anti- (IL-4, TGF, IFN-gamma) inflammatory cytokines and MMP2 and 9 was quantified by real-time PCR, MMP proteins by Western analysis, and MMP activity by gel zymography. RESULTS: Chronic hypoxia increased (P < .05) TNF-alpha, IL-6, MMP2, and MMP9 mRNA levels but not IL-4, TGF, or IFN-gamma. Hypoxia increased protein levels of MMP9 but not MMP2, despite a hypoxia-induced increase in MMP2 activity. CONCLUSION: Intrauterine hypoxia may be an important stimulus in local generation of selected proinflammatory cytokines and MMPs in fetal hearts.


Assuntos
Citocinas/biossíntese , Hipóxia Fetal/complicações , Coração/embriologia , Metaloproteinases da Matriz/biossíntese , Animais , Western Blotting , Feminino , Cobaias , Interferon gama/biossíntese , Interleucina-1beta/biossíntese , Interleucina-4/biossíntese , Interleucina-6/biossíntese , Metaloproteinase 2 da Matriz/biossíntese , Metaloproteinase 9 da Matriz/biossíntese , Gravidez , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fatores de Crescimento Transformadores , Fator de Necrose Tumoral alfa/biossíntese , Regulação para Cima
15.
Early Hum Dev ; 84(2): 121-7, 2008 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17512683

RESUMO

OBJECTIVE: Glutathione is a natural antioxidant in the fetus and adult. We sought to determine whether maternal hypoxia alters glutathione levels in fetal organs as an adaptive response to the reduced oxygenation. STUDY DESIGN: Timed pregnant guinea pigs were housed in either a Plexiglas chamber containing 10.5% O(2) from 46 to 60 days gestation (HPX, n=6) or in room air, as the normoxic control (NMX, n=5). Pregnant guinea pigs were anesthetized at near term ( approximately 60 days, term=65 days) and liver, lungand kidney were excised from anesthetized fetuses and stored frozen (-80 degrees C) prior to sample processing. Using the hypoxia marker, pimonidazole, we measured a hypoxia-induced increase in stained cells of fetal liver compared to no change in either the lung or kidney. To measure the effect of hypoxia among different organs, total glutathione (GSH) content and protein levels of gamma-glutamyl cysteine synthetase (gamma-GCS) were measured from the same organs. RESULTS: Maternal hypoxia increased (P<0.05) total glutathione levels by 121% in the fetal liver but had no effect in either fetal lung or kidney. Chronic hypoxia increased (P<0.05) gamma-GCS protein levels in all three fetal organs studied. CONCLUSION: These results demonstrate that the fetal response to maternal hypoxia may be organ specific. The increase in fetal liver glutathione via upregulation of gamma-GCS may be an important adaptive response to prolonged hypoxic stress.


Assuntos
Hipóxia Fetal/metabolismo , Glutamato-Cisteína Ligase/biossíntese , Glutationa/metabolismo , Fígado/metabolismo , Animais , Western Blotting , Peso Corporal/fisiologia , Feminino , Hipóxia Fetal/sangue , Hipóxia Fetal/enzimologia , Feto , Cobaias , Imuno-Histoquímica , Rim/enzimologia , Rim/metabolismo , Ácido Láctico/sangue , Fígado/embriologia , Fígado/enzimologia , Pulmão/enzimologia , Pulmão/metabolismo , Tamanho do Órgão/fisiologia , Gravidez , Ácido Pirúvico/sangue
16.
J Soc Gynecol Investig ; 11(8): 511-20, 2004 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-15582495

RESUMO

OBJECTIVE: Chronic hypoxia causes redistribution of fetal cardiac output by mechanisms poorly understood. We tested the hypothesis that chronic hypoxia alters vascular reactivity of arteries from near-term fetal guinea pigs. METHODS: Pregnant guinea pigs (50 days, term = 65 days) were exposed to either normoxia (room air) or hypoxia (12% O2) for 14 days. Carotid artery ring segments from anesthetized fetuses were mounted onto myographs for measurement of force. Contractile responses to cumulative addition of prostaglandin F2alpha (PGF2alpha, 10(-9) M to 10(-5) M), U46619, a thromboxane mimetic (10(-12) M to 12(-6) M), and KCl (10 to 120 mM) were measured in the presence and absence of INDO (INDO, 10(-5) M) alone and INDO plus nitro-L-arginine (LNA, 10(-4) M), or INDO plus N6-iminoethyl-L-lysine (LNIL, 5 x 10(-5) M, a selective iNOS inhibitor), and measured in endothelium-intact and denuded arteries. Nitric oxide synthase (NOS) activity was measured in isolated arteries by 14C-L-arginine to 14C-L-citrulline conversion. RESULTS: Hypoxia decreased contractile responses to both PGF2alpha and U46619 under control conditions. Maximal contraction to both agonists was increased in hypoxemic arteries after INDO alone and INDO + LNA compared to normoxic controls. Endothelium-denudation abolished the differences between the groups. KCl contraction was unaffected by hypoxia. LNIL potentiated maximal PGF(2alpha) contraction but was similar between groups. Hypoxia increased (P < .05) total and Ca(2+)-dependent NOS activities by 1.7- and 2.1-fold, respectively, but had no effect on Ca(2+)-independent activity. CONCLUSION: Chronic hypoxia alters vascular reactivity of fetal carotid arteries by increasing the contribution of both vasodilator prostaglandins and nitric oxide and suggests that changes in local vascular mechanisms may be altered by chronic hypoxia.


Assuntos
Artérias/embriologia , Endotélio Vascular/metabolismo , Hipóxia/fisiopatologia , Contração Muscular/efeitos dos fármacos , Óxido Nítrico/biossíntese , Prostaglandinas/biossíntese , Ácido 15-Hidroxi-11 alfa,9 alfa-(epoximetano)prosta-5,13-dienoico/farmacologia , Animais , Artérias/fisiopatologia , Cálcio/farmacologia , Artérias Carótidas/embriologia , Artérias Carótidas/fisiopatologia , Dinoprosta/farmacologia , Feminino , Peso Fetal , Cobaias , Músculo Liso Vascular/embriologia , Óxido Nítrico Sintase/metabolismo , Cloreto de Potássio/farmacologia , Gravidez , Vasoconstritores/farmacologia
17.
High Alt Med Biol ; 4(2): 215-24, 2003.
Artigo em Inglês | MEDLINE | ID: mdl-12855053

RESUMO

This review examines the effect of high altitude and/or chronic hypoxia on cardiac mechanisms that influence perfusion of the fetal heart (e.g., tissue metabolism, coronary vessel growth, and coronary blood flow and vessel responsiveness). In response to intrauterine hypoxia, the fetal heart may either reduce its energy demand or increase its substrate and oxygen delivery as a means of sustaining cardiac function. Cardiac glycolysis predominates as a metabolic pathway of ATP synthesis in the fetal heart under both normoxic and hypoxic conditions. During prolonged oxygen insufficiency, normal cardiac function is sustained by anaerobic glycolysis relying primarily on high levels of stored glycogen in the heart. Chronic hypoxia increases coronary vessel growth and myocardial vascularization in fetal hearts, although the response may depend on the presence of ventricular hypertrophy. Recent studies demonstrate that high altitude hypoxia increases both resting fetal coronary flow and coronary flow reserve as an adaptive response toward increasing oxygen delivery. Hypoxia may also directly effect local vascular smooth muscle mechanisms, resulting in altered coronary artery reactivity to circulating vasoactive substances and contributing to enhanced perfusion. Further study is needed to understand the relative importance of each of these cardiac adaptations in contributing to fetal survival. It is likely that differences in fetal coronary responses to intrauterine hypoxia are highly dependent on the gestational age and relative maturity of the animal species.


Assuntos
Coração/embriologia , Hipóxia/embriologia , Adaptação Fisiológica/fisiologia , Velocidade do Fluxo Sanguíneo/fisiologia , Doença Crônica , Circulação Coronária/fisiologia , Vasos Coronários/embriologia , Vasos Coronários/fisiopatologia , Metabolismo Energético/fisiologia , Coração/fisiopatologia , Humanos , Hipóxia/fisiopatologia
18.
Reprod Sci ; 21(7): 883-891, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24406790

RESUMO

Prenatal hypoxia (HPX) reduces mitochondrial cytochrome c oxidase (CCO and COX) activity in fetal guinea pig (GP) hearts. The aim of this study was to quantify the lasting effects of chronic prenatal HPX on cardiac mitochondrial enzyme activity and protein expression in offspring hearts. Pregnant GPs were exposed to either normoxia (NMX) or HPX (10.5%O2) during the last 14 days of pregnancy. Both NMX and HPX fetuses, delivered vaginally, were housed under NMX conditions until 90 days of age. Total RNA and mitochondrial fractions were isolated from hearts of anesthetized NMX and HPX offspring and showed decreased levels of CCO but not medium-chain acyl dehydrogenase activity, protein levels of nuclear- and mitochondrial-encoded COX4 and COX1, respectively, and messenger RNA expression of peroxisome proliferator-activated receptor gamma coactivator 1-alpha, COX5b, and 4.1 compared to NMX controls. Prenatal HPX may alter mitochondrial function in the offspring by disrupting protein expression associated with the respiratory chain.

19.
Reprod Sci ; 20(3): 299-307, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22923417

RESUMO

We hypothesized that chronic hypoxia disrupts mitochondrial function via oxidative stress in fetal organs. Pregnant guinea pig sows were exposed to either normoxia or hypoxia (10.5% O2, 14 days) in the presence or absence of the antioxidant, N-acetylcysteine (NAC). Near-term anesthetized fetuses were delivered via hysterotomy, and fetal livers, hearts, lungs, and forebrains harvested. We quantified the effects of chronic hypoxia on cytochrome oxidase (CCO) activity and 2 factors known to regulate CCO activity: malondialdehyde (MDA) and CCO subunit 4 (COX4). Hypoxia increased the MDA levels in fetal liver, heart, and lung with a corresponding reduction in CCO activity, prevented by prenatal NAC. The COX4 expression paralleled CCO activity in fetal liver and lung, but was unaltered in fetal hearts due to hypoxia. Hypoxia reduced the brain COX4 expression despite having no effect on CCO activity. This study identifies the mitochondrion as an important target site in tissue-specific oxidative stress for the induction of fetal hypoxic injury.


Assuntos
Complexo IV da Cadeia de Transporte de Elétrons/metabolismo , Coração Fetal/enzimologia , Hipóxia/enzimologia , Fígado/enzimologia , Pulmão/enzimologia , Estresse Oxidativo/fisiologia , Proteínas de Saccharomyces cerevisiae/metabolismo , Animais , Doença Crônica , Ativação Enzimática/fisiologia , Feminino , Coração Fetal/embriologia , Cobaias , Fígado/embriologia , Pulmão/embriologia , Gravidez
20.
Neurotox Res ; 24(1): 80-93, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23192463

RESUMO

In utero exposure to cigarette smoke has severe consequences for the developing fetus, including increased risk of birth complications and behavioral and learning disabilities later in life. Evidence from animal models suggests that the cognitive deficits may be a consequence of in utero nicotine exposure in the brain during critical developmental periods. However, maternal smoking exposes the fetus to not only nicotine but also a hypoxic intrauterine environment. Thus, both nicotine and hypoxia are capable of initiating cellular cascades, leading to long-term changes in synaptic patterning that have the potential to affect cognitive functions. This study investigates the combined effect of in utero exposure to nicotine and hypoxia on neuronal and glial elements in the hippocampal CA1 field. Fetal guinea pigs were exposed in utero to normoxic or hypoxic conditions in the presence or absence of nicotine. Hypoxia increased the protein levels of matrix metalloproteinase-9 (MMP-9) and synaptophysin and decreased the neural density as measured by NeuN immunoreactivity (ir). Nicotine exposure had no effect on these neuronal parameters but dramatically increased the density of astrocytes immunopositive for glial fibrillary acidic protein (GFAP). Further investigation into the effects of in utero nicotine exposure revealed that both GFAP-ir and NeuN-ir in the CA1 field were significantly reduced in adulthood. Taken together, our data suggest that prenatal exposure to nicotine and hypoxia not only alters synaptic patterning acutely during fetal development, but that nicotine also has long-term consequences that are observed well into adulthood. Moreover, these effects most likely take place through distinct mechanisms.


Assuntos
Astrócitos/efeitos dos fármacos , Astrócitos/metabolismo , Giro Denteado/efeitos dos fármacos , Giro Denteado/metabolismo , Feto/efeitos dos fármacos , Nicotina/toxicidade , Efeitos Tardios da Exposição Pré-Natal/induzido quimicamente , Animais , Astrócitos/citologia , Contagem de Células , Giro Denteado/citologia , Feminino , Feto/metabolismo , Cobaias , Hipóxia/metabolismo , Masculino , Metaloproteinase 9 da Matriz/metabolismo , Neurônios/metabolismo , Gravidez , Efeitos Tardios da Exposição Pré-Natal/metabolismo , Sinaptofisina/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA