Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 77
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
J Physiol ; 599(4): 1307-1318, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33347615

RESUMO

KEY POINTS: Chronic hypoxaemia is associated with intrauterine growth restriction (IUGR) and a predisposition to the development of hypertension in adult life. IUGR fetuses exhibit a greater reliance on α-adrenergic activation for blood pressure regulation. The fetal blood pressure response to post-ganglionic blockade is not different between control and IUGR fetuses. The decrease in mean arterial pressure is greater in the IUGR sheep fetus after α-adrenergic receptor blockade at the level of the vasculature and this is inversely related to fetal PO2 . The increased reliance that the IUGR fetus has on α-adrenergic activation for maintenance of mean arterial pressure is not a result of increased post-ganglionic sympathetic activation. ABSTRACT: Intrauterine growth restriction (IUGR) is associated with an increased risk of cardiovascular disease in adult life. Placental restriction (PR) in sheep results in chronic hypoxaemia and early onset IUGR with increased circulating plasma noradrenaline concentrations. These IUGR fetuses exhibit a greater decrease in mean arterial pressure (MAP) during α-adrenergic blockade. We aimed to determine the role of post-ganglionic sympathetic activation with respect to regulating MAP in IUGR fetal sheep. PR was induced by carunclectomy surgery prior to conception. Fetal vascular catheterization was performed at 110-126 days gestational age (GA) (term, 150 days) in nine control and seven PR-IUGR fetuses. The fetal blood pressure response to both a post-ganglionic and an α-adrenergic receptor blocker was assessed at 116-120 days GA and/or 129-131 days GA. The effect of both post ganglionic and α-adrenergic blockade on fetal blood pressure was then compared between control and IUGR fetuses at both GAs. There was no difference in the effect of post-ganglionic blockade on MAP in control and IUGR fetal sheep at either 116-120 days GA or 129-131 days GA. α-adrenergic receptor blockade decreased MAP to the same extent in both control and IUGR fetuses at 116-120 days GA. At 129-131 days GA, the drop in MAP in response to α-adrenergic receptor blockade was greater in IUGR fetuses than controls. There was a significant inverse relationship between the drop in MAP in response to α-adrenergic receptor blockade at both GAs with fetal PO2 . Thus, the increased dependence on α-adrenergic activation for blood pressure regulation in the chronically hypoxaemic IUGR fetus is not a result of increased post-ganglionic sympathetic activation.


Assuntos
Retardo do Crescimento Fetal , Feto , Animais , Pressão Sanguínea , Feminino , Hipóxia , Gravidez , Receptores Adrenérgicos alfa , Ovinos
2.
Reprod Fertil Dev ; 32(9): 835-850, 2020 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-32527374

RESUMO

This study evaluated the effect of protein restriction during the periconception (PERI) and first trimester (POST) periods on maternal performance, physiology and early fetal growth. Yearling nulliparous heifers (n=360) were individually fed a diet high or low in protein (HPeri and LPeri respectively) beginning 60 days before conception. From 24 to 98 days post-conception (dpc), half of each treatment group changed to the alternative post-conception high- or low-protein diet (HPost and LPost respectively), yielding four groups in a 2×2 factorial design with a common diet until parturition. Protein restriction was associated with lower bodyweight subsequent to reduced (but positive) average daily weight gain (ADG) during the PERI and POST periods. During the POST period, ADG was greater in LPeri than HPeri heifers and tended to be greater in LPost than HPost heifers during the second and third trimester. Bodyweight was similar at term. The pregnancy rate did not differ, but embryo loss between 23 and 36 dpc tended to be greater in LPeri than HPeri heifers. Overall, a greater proportion of male fetuses was detected (at 60 dpc 63.3% male vs 36.7% female). Protein restriction altered maternal plasma urea, non-esterified fatty acids, progesterone, leptin and insulin-like growth factor 1 at critical stages of fetal development. However, profiles varied depending on the sex of the conceptus.


Assuntos
Fenômenos Fisiológicos da Nutrição Animal , Dieta com Restrição de Proteínas/veterinária , Fertilização , Desenvolvimento Fetal , Fenômenos Fisiológicos da Nutrição Materna , Técnicas de Reprodução Assistida/veterinária , Ração Animal , Animais , Biomarcadores/sangue , Bovinos , Metabolismo Energético , Feminino , Idade Gestacional , Ganho de Peso na Gestação , Masculino , Gravidez , Taxa de Gravidez , Fatores Sexuais , Razão de Masculinidade
3.
J Physiol ; 596(12): 2345-2358, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29604078

RESUMO

KEY POINTS: This study investigates the impact of decreased fetal plasma glucose concentrations on the developing heart in late gestation, by subjecting pregnant ewes to a 50% global nutrient restriction. Late gestation undernutrition (LGUN) decreased fetal plasma glucose concentrations whilst maintaining a normoxemic blood gas status. LGUN increased the mRNA expression of IGF2 and IGF2R. Fetal plasma glucose concentrations, but not fetal blood pressure, were significantly correlated with IGF2 expression and the activation of CAMKII in the fetal right ventricle. LGUN increased interstitial collagen deposition and altered the protein abundance of phospho-PLB and phospho-troponin I, regulators of cardiac contractility and relaxation. This study shows that a decrease in fetal plasma glucose concentrations may play a role in the development of detrimental changes in the right ventricle in early life, highlighting CAMKII as a potential target for the development of intervention strategies. ABSTRACT: Exposure of the fetus to a range of environmental stressors, including maternal undernutrition, is associated with an increased risk of death from cardiovascular disease in adult life. This study aimed to determine the effect of maternal nutrient restriction in late gestation on the molecular mechanisms that regulate cardiac growth and development of the fetal heart. Maternal undernutrition resulted in a decrease in fetal glucose concentrations across late gestation, whilst fetal arterial PO2 remained unchanged between the control and late gestation undernutrition (LGUN) groups. There was evidence of an up-regulation of IGF2/IGF2R signalling through the CAMKII pathway in the fetal right ventricle in the LGUN group, suggesting an increase in hypertrophic signalling. LGUN also resulted in an increased mRNA expression of COL1A, TIMP1 and TIMP3 in the right ventricle of the fetal heart. In addition, there was an inverse relationship between fetal glucose concentrations and COL1A expression. The presence of interstitial fibrosis in the heart of the LGUN group was confirmed through the quantification of picrosirius red-stained sections of the right ventricle. We have therefore shown that maternal undernutrition in late gestation may drive the onset of myocardial remodelling in the fetal right ventricle and thus has negative implications for right ventricle function and cardiac health in later life.


Assuntos
Colágeno/metabolismo , Doenças Fetais/fisiopatologia , Coração Fetal/fisiopatologia , Desnutrição/complicações , Doenças dos Ovinos/fisiopatologia , Transdução de Sinais , Animais , Feminino , Doenças Fetais/etiologia , Coração Fetal/metabolismo , Fibrose/etiologia , Fibrose/fisiopatologia , Idade Gestacional , Ventrículos do Coração/metabolismo , Ventrículos do Coração/fisiopatologia , Fator de Crescimento Insulin-Like II/metabolismo , Desnutrição/metabolismo , Fenômenos Fisiológicos da Nutrição Materna , Troca Materno-Fetal , Gravidez , Ovinos
4.
Am J Physiol Regul Integr Comp Physiol ; 315(6): R1123-R1153, 2018 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-30325659

RESUMO

Experimental studies that are relevant to human pregnancy rely on the selection of appropriate animal models as an important element in experimental design. Consideration of the strengths and weaknesses of any animal model of human disease is fundamental to effective and meaningful translation of preclinical research. Studies in sheep have made significant contributions to our understanding of the normal and abnormal development of the fetus. As a model of human pregnancy, studies in sheep have enabled scientists and clinicians to answer questions about the etiology and treatment of poor maternal, placental, and fetal health and to provide an evidence base for translation of interventions to the clinic. The aim of this review is to highlight the advances in perinatal human medicine that have been achieved following translation of research using the pregnant sheep and fetus.


Assuntos
Feto/metabolismo , Placenta/metabolismo , Resultado da Gravidez , Ovinos/fisiologia , Animais , Modelos Animais de Doenças , Feminino , Humanos , Troca Materno-Fetal/fisiologia , Gravidez , Prenhez
5.
Pharmacol Res ; 134: 68-78, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-29890254

RESUMO

The effects of intrauterine growth restriction (IUGR) extend well into postnatal life. IUGR is associated with an increased risk of adverse health outcomes, which often leads to greater medication usage. Many medications require hepatic metabolism for activation or clearance, but hepatic function may be altered in IUGR fetuses. Using a sheep model of IUGR, we determined the impact of IUGR on hepatic drug metabolism and drug transporter expression, both important mediators of fetal drug exposure, in late gestation and in neonatal life. In the late gestation fetus, IUGR decreased the gene expression of uptake drug transporter OATPC and increased P-glycoprotein protein expression in the liver, but there was no change in the activity of the drug metabolising enzymes CYP3A4 or CYP2D6. In contrast, at 3 weeks of age, CYP3A4 activity was reduced in the livers of lambs born with low birth weight (LBW), indicating that LBW results in changes to drug metabolising capacity in neonatal life. Together, these results suggest that IUGR may reduce hepatic drug metabolism in fetal and neonatal life through different mechanisms.


Assuntos
Membro 1 da Subfamília B de Cassetes de Ligação de ATP/metabolismo , Citocromo P-450 CYP2D6/metabolismo , Citocromo P-450 CYP3A/metabolismo , Retardo do Crescimento Fetal/enzimologia , Fígado/enzimologia , Transportadores de Ânions Orgânicos/metabolismo , Preparações Farmacêuticas/metabolismo , Membro 1 da Subfamília B de Cassetes de Ligação de ATP/genética , Animais , Animais Recém-Nascidos , Peso ao Nascer , Citocromo P-450 CYP2D6/genética , Citocromo P-450 CYP3A/genética , Modelos Animais de Doenças , Feminino , Retardo do Crescimento Fetal/genética , Peso Fetal , Regulação da Expressão Gênica no Desenvolvimento , Regulação Enzimológica da Expressão Gênica , Idade Gestacional , Transportadores de Ânions Orgânicos/genética , Gravidez , Carneiro Doméstico
6.
J Physiol ; 595(21): 6635-6652, 2017 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-28759122

RESUMO

KEY POINTS: Offspring of overweight and obese women are at greater risk for respiratory complications at birth. We determined the effect of late gestation maternal overnutrition (LGON) in sheep on surfactant maturation, glucose transport and fatty acid metabolism in the lung in fetal and postnatal life. There were significant decreases in surfactant components and numerical density of surfactant producing cells in the alveolar epithelium due to LGON in the fetal lung. However, there were no differences in the levels of these surfactant components between control and LGON lambs at 30 days of age. The reduced capacity for surfactant production in fetuses as a result of LGON may affect the transition to air breathing at birth. There was altered glucose transport and fatty acid metabolism in the lung as a result of LGON in postnatal life. However, there is a normalisation of surfactant components that suggests accelerated maturation in the lungs after birth. ABSTRACT: With the increasing incidence of obesity worldwide, the proportion of women entering pregnancy overweight or obese has increased dramatically. The fetus of an overnourished mother experiences numerous metabolic changes that may modulate lung development and hence successful transition to air breathing at birth. We used a sheep model of maternal late gestation overnutrition (LGON; from 115 days' gestation, term 147 ± 3 days) to determine the effect of exposure to an increased plane of nutrition in late gestation on lung development in the fetus (at 141 days' gestation) and the lamb (30 days after birth). We found a decrease in the numerical density of surfactant protein positive cells, as well as a reduction in mRNA expression of surfactant proteins (SFTP-A, -B and -C), a rate limiting enzyme in surfactant phospholipid synthesis (phosphate cytidylyltransferase 1, choline, α; PCYT1A), and glucose transporters (SLC2A1 and SLC2A4) in the fetal lung. In lambs at 30 days after birth, there were no differences between Control and LGON groups in the surfactant components that were downregulated in the LGON fetuses. However, mRNA expression of SFTP-A, PCYT1A, peroxisome proliferator activated receptor-γ, fatty acid synthase and fatty acid transport protein were increased in LGON lambs compared to controls. These results indicate a reduced capacity for surfactant production in late gestation. While these deficits are normalised by 30 days after birth, the lungs of LGON lambs exhibited altered glucose transport and fatty acid metabolism, which is consistent with an enhanced capacity for surfactant synthesis and restoration of surfactant maturity in these animals.


Assuntos
Pulmão/embriologia , Hipernutrição/metabolismo , Complicações na Gravidez/metabolismo , Efeitos Tardios da Exposição Pré-Natal/metabolismo , Proteínas Associadas a Surfactantes Pulmonares/metabolismo , Animais , Ácidos Graxos/metabolismo , Feminino , Glucose/metabolismo , Pulmão/metabolismo , Pulmão/patologia , Hipernutrição/patologia , Gravidez , Complicações na Gravidez/patologia , Efeitos Tardios da Exposição Pré-Natal/patologia , Proteínas Associadas a Surfactantes Pulmonares/genética , Mucosa Respiratória/embriologia , Mucosa Respiratória/metabolismo , Mucosa Respiratória/patologia , Ovinos
7.
Am J Physiol Endocrinol Metab ; 310(2): E148-59, 2016 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-26487010

RESUMO

We have investigated the effects of embryo number and maternal undernutrition imposed either around the time of conception or before implantation on hepatic lipid metabolism in the sheep fetus. We have demonstrated that periconceptional undernutrition and preimplantation undernutrition each resulted in decreased hepatic fatty acid ß-oxidation regulators, PGC-1α (P < 0.05), PDK2 (P < 0.01), and PDK4 (P < 0.01) mRNA expression in singleton and twin fetuses at 135-138 days gestation. In singletons, there was also lower hepatic PDK4 (P < 0.01), CPT-1 (P < 0.01), and PKCζ (P < 0.01) protein abundance in the PCUN and PIUN groups and a lower protein abundance of PDPK-1 (P < 0.05) in the PCUN group. Interestingly, in twins, the hepatic protein abundance of p-AMPK (Ser(485)) (P < 0.01), p-PDPK-1 (Ser(41)) (P < 0.05), and PKCζ (P < 0.05) was higher in the PCUN and PIUN groups, and hepatic PDK4 (P < 0.001) and CPT-1 (P < 0.05) protein abundance was also higher in the PIUN twin fetus. We also found that the expression of a number of microRNAs was altered in response to PCUN or PIUN and that there is evidence that these changes may underlie the changes in the protein abundance of key regulators of hepatic fatty acid ß-oxidation in the PCUN and PIUN groups. Therefore, embryo number and the timing of maternal undernutrition in early pregnancy have a differential impact on hepatic microRNA expression and on the factors that regulate hepatic fatty acid oxidation and lipid synthesis.


Assuntos
Metabolismo dos Lipídeos/fisiologia , Fígado/metabolismo , Desnutrição/metabolismo , Fenômenos Fisiológicos da Nutrição Materna/fisiologia , MicroRNAs/metabolismo , Animais , Feminino , Fertilização/fisiologia , Regulação da Expressão Gênica no Desenvolvimento , MicroRNAs/genética , Gravidez , Proteínas Quinases/genética , Proteínas Quinases/metabolismo , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/metabolismo , Ovinos , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
8.
BMC Genomics ; 16: 541, 2015 Jul 22.
Artigo em Inglês | MEDLINE | ID: mdl-26198574

RESUMO

BACKGROUND: There is a limited capacity to repair damage in the mammalian heart after birth, which is primarily due to the inability of cardiomyocytes to proliferate after birth. This is in contrast to zebrafish and salamander, in which cardiomyocytes retain the ability to proliferate throughout life and can regenerate their heart after significant damage. Recent studies in zebrafish and rodents implicate microRNA (miRNA) in the regulation of genes responsible for cardiac cell cycle progression and regeneration, in particular, miR-133a, the miR-15 family, miR-199a and miR-590. However, the significance of these miRNA and miRNA in general in the regulation of cardiomyocyte proliferation in large mammals, including humans, where the timing of heart development relative to birth is very different than in rodents, is unclear. To determine the involvement of miRNA in the down-regulation of cardiomyocyte proliferation occurring before birth in large mammals, we investigated miRNA and target gene expression in sheep hearts before and after birth. The experimental approach included targeted transcriptional profiling of miRNA and target mRNA previously identified in rodent studies as well as genome-wide miRNA profiling using microarrays. RESULTS: The cardiac expression of miR-133a increased and its target gene IGF1R decreased with increasing age, reaching their respective maximum and minimum abundance when the majority of ovine cardiomyocytes were quiescent. The expression of the miR-15 family members was variable with age, however, four of their target genes decreased with age. These latter profiles are inconsistent with the direct involvement of this family of miRNA in cardiomyocyte quiescence in late gestation sheep. The expression patterns of 'pro-proliferative' miR-199a and miR-590 were also inconsistent with their involvement in cardiomyocyte quiescence. Consequently, miRNA microarray analysis was undertaken, which identified six discrete clusters of miRNA with characteristic developmental profiles. The functions of predicted target genes for the miRNA in four of the six clusters were enriched for aspects of cell division and regulation of cell proliferation suggesting a potential role of these miRNA in regulating cardiomyocyte proliferation. CONCLUSION: The results of this study show that the expression of miR-133a and one of its target genes is consistent with it being involved in the suppression of cardiomyocyte proliferation, which occurs across the last third of gestation in sheep. The expression patterns of the miR-15 family, miR-199a and miR-590 were inconsistent with direct involvement in the regulation cardiomyocyte proliferation in sheep, despite studies in rodents demonstrating that their manipulation can influence the degree of cardiomyocyte proliferation. miRNA microarray analysis suggests a coordinated and potentially more complex role of multiple miRNA in the regulation of cardiomyocyte quiescence and highlights significant differences between species that may reflect their substantial differences in the timing of this developmental process.


Assuntos
Coração/crescimento & desenvolvimento , MicroRNAs/genética , Miócitos Cardíacos/fisiologia , Ovinos/genética , Animais , Proliferação de Células/genética , Perfilação da Expressão Gênica , Regulação da Expressão Gênica no Desenvolvimento , Humanos , MicroRNAs/biossíntese , Análise em Microsséries , RNA Mensageiro/genética , Ovinos/crescimento & desenvolvimento
9.
Am J Physiol Lung Cell Mol Physiol ; 309(1): L84-97, 2015 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-25934670

RESUMO

Experimental placental restriction (PR) by carunclectomy in fetal sheep results in intrauterine growth restriction (IUGR), chronic hypoxemia, increased plasma cortisol, and decreased lung surfactant protein (SP) expression. The mechanisms responsible for decreased SP expression are unknown but may involve decreased glucocorticoid (GC) action or changes in hypoxia signaling. Endometrial caruncles were removed from nonpregnant ewes to induce PR. Lungs were collected from control and PR fetuses at 130-135 (n = 19) and 139-145 (n = 28) days of gestation. qRT-PCR and Western blotting were used to quantify lung mRNA and protein expression, respectively, of molecular regulators and downstream targets of the GC and hypoxia-signaling pathways. We confirmed a decrease in SP-A, -B, and -C, but not SP-D, mRNA expression in PR fetuses at both ages. There was a net downregulation of GC signaling with a reduction in GC receptor (GR)-α and -ß protein expression and a decrease in the cofactor, GATA-6. GC-responsive genes including transforming growth factor-ß1, IL-1ß, and ß2-adrenergic receptor were not stimulated. Prolyl hydroxylase domain (PHD)2 mRNA and protein and PHD3 mRNA expression increased with a concomitant increase in hypoxia-inducible factor-1α (HIF-1α) and HIF-1ß mRNA expression. There was an increase in mRNA expression of several, but not all, hypoxia-responsive genes. Hence, both GC and hypoxia signaling may contribute to reduced SP expression. Although acute hypoxia normally inactivates PHDs, chronic hypoxemia in the PR fetus increased PHD abundance, which normally prevents HIF signaling. This may represent a mechanism by which chronic hypoxemia contributes to the decrease in SP production in the IUGR fetal lung.


Assuntos
Retardo do Crescimento Fetal/patologia , Hipóxia Fetal/patologia , Pulmão/embriologia , Prolil Hidroxilases/metabolismo , Proteínas Associadas a Surfactantes Pulmonares/metabolismo , Surfactantes Pulmonares/metabolismo , Animais , Desenvolvimento Fetal , Retardo do Crescimento Fetal/metabolismo , Fator de Transcrição GATA6/metabolismo , Glucocorticoides/metabolismo , Hidrocortisona/sangue , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Prolina Dioxigenases do Fator Induzível por Hipóxia/metabolismo , Interleucina-1beta/metabolismo , Pulmão/enzimologia , Prolil Hidroxilases/biossíntese , Estrutura Terciária de Proteína , Proteína D Associada a Surfactante Pulmonar/metabolismo , RNA Mensageiro/genética , Receptores Adrenérgicos/metabolismo , Receptores de Glucocorticoides/metabolismo , Ovinos/genética , Transdução de Sinais , Fator de Crescimento Transformador beta1/metabolismo
10.
Am J Physiol Regul Integr Comp Physiol ; 308(3): R151-62, 2015 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-25427766

RESUMO

In the fetus, there is a redistribution of cardiac output in response to acute hypoxemia, to maintain perfusion of key organs, including the brain, heart, and adrenal glands. There may be a similar redistribution of cardiac output in the chronically hypoxemic, intrauterine growth-restricted fetus. Surgical removal of uterine caruncles in nonpregnant ewe results in the restriction of placental growth (PR) and intrauterine growth. Vascular catheters were implanted in seven control and six PR fetal sheep, and blood flow to organs was determined using microspheres. Placental and fetal weight was significantly reduced in the PR group. Despite an increase in the relative brain weight in the PR group, there was no difference in blood flow to the brain between the groups, although PR fetuses had higher blood flow to the temporal lobe. Adrenal blood flow was significantly higher in PR fetuses, and there was a direct relationship between mean gestational PaO2 and blood flow to the adrenal gland. There was no change in blood flow, but a decrease in oxygen and glucose delivery to the heart in the PR fetuses. In another group, there was a decrease in femoral artery blood flow in the PR compared with the Control group, and this may support blood flow changes to the adrenal and temporal lobe. In contrast to the response to acute hypoxemia, these data show that there is a redistribution of blood flow to the adrenals and temporal lobe, but not the heart or whole brain, in chronically hypoxemic PR fetuses in late gestation.


Assuntos
Glândulas Suprarrenais/irrigação sanguínea , Encéfalo/irrigação sanguínea , Vasos Coronários/crescimento & desenvolvimento , Retardo do Crescimento Fetal/fisiopatologia , Hipóxia/sangue , Efeitos Tardios da Exposição Pré-Natal , Animais , Feminino , Retardo do Crescimento Fetal/sangue , Feto/fisiologia , Idade Gestacional , Coração/fisiopatologia , Placenta/irrigação sanguínea , Gravidez , Ovinos
11.
Am J Physiol Regul Integr Comp Physiol ; 308(7): R627-35, 2015 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-25632020

RESUMO

The cardiac insulin-like growth factor 2 receptor (IGF-2R) can induce cardiomyocyte hypertrophy in a heterotrimeric G protein receptor-coupled manner involving αq (Gαq) or αs (Gαs). We have previously shown increased left ventricular weight and cardiac IGF-2 and IGF-2R gene expression in low-birth-weight (LBW) compared with average-birth-weight (ABW) lambs. Here, we have investigated the cardiac expression of IGF-2 gene variants, the degree of histone acetylation, and the abundance of proteins in the IGF-2R downstream signaling pathway in ABW and LBW lambs. Samples from the left ventricle of ABW and LBW lambs were collected at 21 days of age. There was increased phospho-CaMKII protein with decreased HDAC 4 abundance in the LBW compared with ABW lambs. There was increased GATA 4 and decreased phospho-troponin I abundance in LBW compared with ABW lambs, which are markers of pathological cardiac hypertrophy and impaired or reduced contractility, respectively. There was increased histone acetylation of H3K9 at IGF-2R promoter and IGF-2R intron 2 differentially methylated region in the LBW lamb. In conclusion, histone acetylation of IGF-2R may lead to increased IGF-2R mRNA expression and subsequently mediate Gαq signaling early in life via CaMKII, resulting in an increased risk of left ventricular hypertrophy and cardiovascular disease in adult life.


Assuntos
Peso ao Nascer , Subunidades alfa Gq-G11 de Proteínas de Ligação ao GTP/metabolismo , Ventrículos do Coração/metabolismo , Hipertrofia Ventricular Esquerda/metabolismo , Miocárdio/metabolismo , Receptor IGF Tipo 2/metabolismo , Transdução de Sinais , Acetilação , Fatores Etários , Animais , Animais Recém-Nascidos , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/metabolismo , Fator de Transcrição GATA4/metabolismo , Regulação da Expressão Gênica , Ventrículos do Coração/patologia , Ventrículos do Coração/fisiopatologia , Histona Desacetilases/metabolismo , Histonas/metabolismo , Hipertrofia Ventricular Esquerda/genética , Hipertrofia Ventricular Esquerda/patologia , Hipertrofia Ventricular Esquerda/fisiopatologia , Contração Miocárdica , Miocárdio/patologia , Fosforilação , Regiões Promotoras Genéticas , RNA Mensageiro/metabolismo , Receptor IGF Tipo 2/genética , Ovinos , Troponina I/metabolismo , Função Ventricular Esquerda
12.
Am J Physiol Endocrinol Metab ; 307(2): E141-50, 2014 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-24844259

RESUMO

Exposure to poor maternal nutrition around the time of conception results in an early prepartum activation of the fetal pituitary-adrenal axis and in increased adrenal growth and stress response after birth associated with epigenetic changes in a differentially methylated region (DMR) of adrenal IGF2/H19. We have determined the effects of maternal undernutrition during the periconceptional period (PCUN: 70% of control intake from 60 days before until 6 days after conception) and early preimplantation period (PIUN: 70% of control intake for 6 days after conception) on fetal plasma ACTH and cortisol concentrations and fetal adrenal ACTHR, StAR, 3ßHSD, CYP11B, CYP17, TGFß1, IGF1, IGF1R, IGF2, and IGF2R mRNA expression and the methylation level of sites within the DMRs of IGF2/H19 and IGF2R in the adrenal of twin and singleton fetuses at 136-138 days gestation. Being a twin resulted in a delayed prepartum increase in fetal ACTH and in a lower cortisol response to CRH in the control but not PCUN and PIUN groups. PCUN, but not PIUN, resulted in an increase in adrenal weight and CYP17 expression in singletons, a decrease in adrenal IGF2 expression in singletons, and an increase in adrenal IGF2R expression in both twins and singletons. IGF2/H19 and IGF2R DMR methylation levels and ACTHR expression were lower in the twin adrenal. Thus, exposure of the oocyte and embryo to maternal undernutrition or to the environment of a twin pregnancy have differential effects on epigenetic and other factors that regulate fetal adrenal growth and IGF2 and IGF2R expression.


Assuntos
Glândulas Suprarrenais/embriologia , Embrião de Mamíferos/embriologia , Epigênese Genética , Desnutrição/fisiopatologia , Fenômenos Fisiológicos da Nutrição Materna , Prenhez/fisiologia , Gravidez Múltipla/fisiologia , Ovinos/embriologia , Glândulas Suprarrenais/metabolismo , Hormônio Adrenocorticotrópico/sangue , Animais , Metilação de DNA , Embrião de Mamíferos/metabolismo , Feminino , Fertilização/fisiologia , Genótipo , Hidrocortisona/sangue , Gravidez , Prenhez/genética , Prenhez/metabolismo , Gravidez Múltipla/genética , Gravidez Múltipla/metabolismo , Ovinos/genética , Ovinos/fisiologia
13.
Am J Physiol Endocrinol Metab ; 306(9): E1013-24, 2014 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-24496309

RESUMO

This study aimed to determine whether exposure of the oocyte and/or embryo to maternal undernutrition results in the later programming of insulin action in the liver and factors regulating gluconeogenesis. To do this, we collect livers from singleton and twin fetal sheep that were exposed to periconceptional (PCUN; -60 to 7 days) or preimplantation (PIUN; 0-7 days) undernutrition at 136-138 days of gestation (term = 150 days). The mRNA and protein abundance of insulin signaling and gluconeogenic factors were then quantified using qRT-PCR and Western blotting, respectively, and global microRNA expression was quantified using deep sequencing methodology. We found that hepatic PEPCK-C mRNA (P < 0.01) and protein abundance and the protein abundance of IRS-1 (P < 0.01), p110ß (P < 0.05), PTEN (P < 0.05), CREB (P < 0.01), and pCREB (Ser(133); P < 0.05) were decreased in the PCUN and PIUN singletons. In contrast, hepatic protein abundance of IRS-1 (P < 0.01), p85 (P < 0.01), p110ß (P < 0.001), PTEN (P < 0.01), Akt2 (P < 0.01), p-Akt (Ser(473); P < 0.01), and p-FOXO-1 (Thr24) (P < 0.01) was increased in twins. There was a decrease in PEPCK-C mRNA (P < 0.01) but, paradoxically, an increase in PEPCK-C protein (P < 0.001) in twins. Both PCUN and PIUN altered the hepatic expression of 23 specific microRNAs. We propose that the differential impact of maternal undernutrition in the presence of one or two embryos on mRNAs and proteins involved in the insulin signaling and gluconeogenesis is explained by changes in the expression of a suite of specific candidate microRNAs.


Assuntos
Gluconeogênese/genética , Insulina/metabolismo , Tamanho da Ninhada de Vivíparos , Fígado/embriologia , Fígado/metabolismo , Desnutrição/metabolismo , MicroRNAs/metabolismo , Animais , Embrião de Mamíferos , Feminino , Fertilização , Feto/metabolismo , Fenômenos Fisiológicos da Nutrição Materna , Gravidez , Carneiro Doméstico , Transdução de Sinais , Fatores de Tempo
14.
Biol Reprod ; 90(1): 5, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24258211

RESUMO

Maternal undernutrition around the time of conception is associated with an increased risk of insulin resistance in adulthood. We determined the effect of maternal undernutrition in the periconceptional period (PCUN, i.e., 60 days prior to 6 days after conception) and the preimplantation period (PIUN, i.e., 0-6 days after conception) on mRNA expression and protein abundance of key insulin-signaling molecules as well as the global microRNA expression in quadriceps muscle of singleton and twin fetal sheep in late gestation. In singleton fetuses, exposure to PCUN resulted in lower protein abundance of PIK3CB (P < 0.01), PRKCZ (P < 0.05), and pPRKCZ (Thr410) (P < 0.05) in skeletal muscle compared to controls. In PIUN singletons, there was a higher protein abundance of IRS1 (P < 0.05), PDPK1 (P < 0.05), and SLC2A4 (P < 0.05) compared to controls. In twins, PCUN resulted in higher protein abundance of IRS1 (P < 0.05), AKT2 (P < 0.05), PDPK1 (P < 0.05), and PRKCZ (P < 0.001), while PIUN also resulted in higher protein abundance of IRS1 (P < 0.05), PRKCZ (P < 0.001), and SLC2A4 (P < 0.05) in fetal muscle compared to controls. There were specific patterns of the types and direction of changes in the expression of 22 microRNAs in skeletal muscle after exposure to PCUN or PIUN and clear differences in these patterns between singleton and twin pregnancies. These findings provide evidence that maternal undernutrition around the time of conception induces changes in the expression of microRNAs, which may play a role in altering the abundance of the key insulin-signaling molecules in skeletal muscle and in the association between PCUN undernutrition and insulin resistance in adult life.


Assuntos
Fertilização , Feto/metabolismo , Insulina/metabolismo , Desnutrição/genética , Fenômenos Fisiológicos da Nutrição Materna , MicroRNAs/genética , Músculo Esquelético/metabolismo , Animais , Desenvolvimento Embrionário/genética , Feminino , Fertilização/fisiologia , Tamanho da Ninhada de Vivíparos , Desnutrição/metabolismo , Fenômenos Fisiológicos da Nutrição Materna/genética , MicroRNAs/metabolismo , Gravidez , Efeitos Tardios da Exposição Pré-Natal/genética , Efeitos Tardios da Exposição Pré-Natal/metabolismo , Carneiro Doméstico , Transdução de Sinais/genética
15.
Am J Physiol Regul Integr Comp Physiol ; 307(5): R538-45, 2014 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-24990855

RESUMO

Increased circulating fetal glucose and insulin concentrations are potential inhibitors of fetal lung maturation and may contribute to the pathogenesis of respiratory distress syndrome (RDS) in infants of diabetic mothers. In this study, we examined the effect of intrafetal glucose infusion on mRNA expression of glucose transporters, insulin-like growth factor signaling, glucocorticoid regulatory genes, and surfactant proteins in the lung of the late-gestation sheep fetus. The numerical density of the cells responsible for producing surfactant was determined using immunohistochemistry. Glucose infusion for 10 days did not affect mRNA expression of glucose transporters or IGFs but did decrease IGF-1R expression. There was reduced mRNA expression of the glucocorticoid-converting enzyme HSD11B-1 and the glucocorticoid receptor, potentially reducing glucocorticoid responsiveness in the fetal lung. Furthermore, surfactant protein (SFTP) mRNA expression was reduced in the lung following glucose infusion, while the number of SFTP-B-positive cells remained unchanged. These findings suggest the presence of a glucocorticoid-mediated mechanism regulating delayed maturation of the surfactant system in the sheep fetus following glucose infusion and provide evidence for the link between abnormal glycemic control during pregnancy and the increased risk of RDS in infants of uncontrolled diabetic mothers.


Assuntos
Feto/metabolismo , Glucocorticoides/metabolismo , Glucose/farmacologia , Pulmão/metabolismo , Surfactantes Pulmonares/metabolismo , RNA Mensageiro/metabolismo , Ovinos/fisiologia , Transdução de Sinais/efeitos dos fármacos , 11-beta-Hidroxiesteroide Desidrogenase Tipo 1/metabolismo , Animais , Gasometria , Peso Corporal/efeitos dos fármacos , Peso Corporal/fisiologia , Feminino , Glucose/metabolismo , Insulina/metabolismo , Pulmão/embriologia , Modelos Animais , Gravidez , Prenhez , Receptor IGF Tipo 1/metabolismo , Receptores de Glucocorticoides/metabolismo , Ovinos/embriologia , Transdução de Sinais/fisiologia
16.
Am J Physiol Regul Integr Comp Physiol ; 306(6): R429-37, 2014 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-24477540

RESUMO

It is unknown whether cardiomyocyte hypertrophy and the transition to fatty acid oxidation as the main source of energy after birth is dependent on the maturation of the cardiomyocytes' metabolic system, or on the limitation of substrate availability before birth. This study aimed to investigate whether intrafetal administration of a peroxisome proliferator-activated receptor-γ (PPAR-γ) agonist, rosiglitazone, during late gestation can stimulate the expression of factors regulating cardiac growth and metabolism in preparation for birth, and the consequences of cardiac contractility in the fetal sheep at ∼140 days gestation. The mRNA expression and protein abundance of key factors regulating growth and metabolism were quantified using quantitative RT-PCR and Western blot analysis, respectively. Cardiac contractility was determined by measuring the Ca(2+) sensitivity and maximum Ca(2+)-activated force of skinned cardiomyocyte bundles. Rosiglitazone-treated fetuses had a lower cardiac abundance of insulin-signaling molecules, including insulin receptor-ß, insulin receptor substrate-1 (IRS-1), phospho-IRS-1 (Tyr-895), phosphatidylinositol 3-kinase (PI3K) regulatory subunit p85, PI3K catalytic subunit p110α, phospho-3-phosphoinositide-dependent protein kinase 1 (Ser-241), protein kinase B (Akt-1), phospho-Akt (Ser-273), PKCζ, phospho-PKCζ(Thr-410), Akt substrate 160 kDa (AS160), phospho-AS160 (Thr-642), and glucose transporter type-4. Additionally, cardiac abundance of regulators of fatty acid ß-oxidation, including adiponectin receptor 1, AMPKα, phospho-AMPKα (Thr-172), phospho-acetyl CoA carboxylase (Ser-79), carnitine palmitoyltransferase-1, and PGC-1α was lower in the rosiglitazone-treated group. Rosiglitazone administration also resulted in a decrease in cardiomyocyte size. Rosiglitazone administration in the late-gestation sheep fetus resulted in a decreased abundance of factors regulating cardiac glucose uptake, fatty acid ß-oxidation, and cardiomyocyte size. These findings suggest that activation of PPAR-γ using rosiglitazone does not promote the maturation of cardiomyocytes; rather, it may decrease cardiac metabolism and compromise cardiac health later in life.


Assuntos
Coração/efeitos dos fármacos , Coração/embriologia , Miócitos Cardíacos/efeitos dos fármacos , PPAR gama/agonistas , Tiazolidinedionas/farmacologia , Animais , Tamanho Celular/efeitos dos fármacos , Ácidos Graxos/metabolismo , Feminino , Feto/efeitos dos fármacos , Feto/metabolismo , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Idade Gestacional , Hipoglicemiantes/farmacologia , Insulina/metabolismo , Contração Miocárdica/efeitos dos fármacos , Contração Miocárdica/fisiologia , Miocárdio/citologia , Miocárdio/metabolismo , Miócitos Cardíacos/citologia , Miócitos Cardíacos/metabolismo , PPAR gama/metabolismo , Gravidez , Proteínas Quinases/genética , Proteínas Quinases/metabolismo , Receptor de Insulina/genética , Receptor de Insulina/metabolismo , Rosiglitazona , Carneiro Doméstico
17.
Pediatr Res ; 75(1-1): 14-21, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24452591

RESUMO

BACKGROUND: Exposure to maternal obesity or hyperglycemia increases the risk of obesity and poor glucose tolerance in the offspring. We hypothesized that maternal overnutrition in late pregnancy would result in (i) lower methylation in the promoter region of the cytosolic form of phosphoenolpyruvate carboxykinase (PEPCK-C; PCK1) and (ii) higher expression of hepatic gluconeogenic factors in the fetal and postnatal lamb. METHODS: Ewes were fed 100% (n = 18) or ~155% (n = 17) of energy requirements from 115 d gestation, and livers were collected at ~140 d gestation or 30 d postnatal age. RESULTS: Maternal overnutrition resulted in a decrease in hepatic expression of the mitochondrial form of PEPCK (PEPCK-M; PCK2) but not of PEPCK-C or glucose-6-phosphatase (G6PHOS) before and after birth. Hepatic expression of peroxisome proliferator-activated receptor γ coactivator 1 (PGC-1), peroxisome proliferator-activated receptor α (PPARα), PEPCK-C, G6PHOS, and 11ß hydroxysteroid dehydrogenase type 1 (11ßHSD1), but not PEPCK-M, was higher in the postnatal lamb compared with that in the fetal lamb. The level of PCK1 methylation was paradoxically approximately twofold higher in the postnatal liver compared with that in the fetal liver. CONCLUSION: Maternal overnutrition programs a decrease in hepatic PEPCK-M in the offspring and as ~50% of total hepatic PEPCK is PEPCK-M, the longer-term consequences of this decrease may be significant.


Assuntos
Metilação de DNA , Gluconeogênese , Fígado/metabolismo , Hipernutrição , Fosfoenolpiruvato Carboxiquinase (ATP)/genética , Regiões Promotoras Genéticas , Animais , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Gluconeogênese/genética , Fígado/embriologia , Fígado/enzimologia , Gravidez , Ovinos/embriologia , Ovinos/crescimento & desenvolvimento
18.
Am J Physiol Endocrinol Metab ; 305(8): E931-41, 2013 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-23921136

RESUMO

Maternal undernutrition around the time of conception is associated with an increased risk of insulin resistance in adulthood. We hypothesized that maternal undernutrition during the periconceptional (PCUN: -60 to 7 days) and/or preimplantation (PIUN: 0-7 days) periods would result in a decrease in UCP1 expression and the abundance of insulin signaling molecules and an increase in the abundance of factors that regulate adipogenesis and lipogenesis in fetal perirenal adipose tissue (PAT) and that these effects would be different in singletons and twins. Maternal PCUN and PIUN resulted in a decrease in UCP1 expression in PAT, and PIUN resulted in higher circulating insulin concentrations, an increased abundance of pPKCζ and PDK4, and a decreased abundance of Akt1, phosphorylated mTOR, and PPARγ in PAT in singleton and twin fetuses. In singletons, there was also a decrease in the abundance of p110ß in PAT in the PCUN and PIUN groups and an increase in total AMPKα in PAT in the PIUN group. In twins, however, there was an increase in the abundance of mTOR in the PCUN group and an increase in PDK2 and decrease in total AMPKα in the PIUN group. Thus exposure to periconceptional undernutrition programs changes in the thermogenic capacity and the insulin and fatty acid oxidation signaling pathway in visceral fat, and these effects are different in singletons and twins. These findings are important, as the thermogenic capacity of brown fat and the insulin sensitivity of visceral fat are important determinants of the risk of developing obesity and an insulin resistance phenotype in later life.


Assuntos
Adipogenia , Desenvolvimento Fetal , Regulação da Expressão Gênica no Desenvolvimento , Gordura Intra-Abdominal/metabolismo , Lipogênese , Desnutrição/fisiopatologia , Fenômenos Fisiológicos da Nutrição Materna , Animais , Animais Endogâmicos , Austrália , Feminino , Fertilização , Hiperinsulinismo/embriologia , Hiperinsulinismo/etiologia , Gordura Intra-Abdominal/embriologia , Canais Iônicos/genética , Canais Iônicos/metabolismo , Tamanho da Ninhada de Vivíparos , Proteínas Mitocondriais/genética , Proteínas Mitocondriais/metabolismo , Gravidez , RNA Mensageiro/metabolismo , Distribuição Aleatória , Carneiro Doméstico , Transdução de Sinais , Proteína Desacopladora 1
19.
Reproduction ; 146(5): 443-54, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24084173

RESUMO

Exposure to dietary restriction during the periconceptional period in either normal or obese ewes results in increased adrenal growth and a greater cortisol response to stress in the offspring, but the mechanisms that programme these changes are not fully understood. Activation of the angiotensin type 1 receptor (AT1R) has been demonstrated to stimulate adrenal growth and steroidogenesis. We have used an embryo transfer model in the sheep to investigate the effects of exposure to dietary restriction in normal or obese mothers from before and 1 week after conception on the methylation status, expression, abundance and localisation of key components of the renin-angiotensin system (RAS) in the adrenal of post-natal lambs. Maternal dietary restriction in normal or obese ewes during the periconceptional period resulted in an increase in angiotensin-converting enzyme (ACE) and AT1R abundance in the absence of changes in the methylation status or mRNA expression of ACE and AT1R in the adrenal of the offspring. Exposure to maternal obesity alone also resulted in an increase in adrenal AT1R abundance. There was no effect of maternal dietary restriction or obesity on ACE2 and AT2R or on ERK, calcium/calmodulin-dependent kinase II abundance, and their phosphorylated forms in the lamb adrenal. Thus, weight loss around the time of conception, in both normal-weight and obese ewes, results in changes within the intra-adrenal RAS consistent with increased AT1R activation. These changes within the intra-adrenal RAS system may contribute to the greater adrenal stress response following exposure to signals of adversity in the periconceptional period.


Assuntos
Glândulas Suprarrenais/metabolismo , Fertilização , Desnutrição/fisiopatologia , Obesidade/dietoterapia , Peptidil Dipeptidase A/biossíntese , Fenômenos Fisiológicos da Nutrição Pré-Natal , Receptor Tipo 1 de Angiotensina/biossíntese , Glândulas Suprarrenais/enzimologia , Glândulas Suprarrenais/crescimento & desenvolvimento , Animais , Animais Endogâmicos , Dieta Redutora/efeitos adversos , Transferência Embrionária , Feminino , Masculino , Desnutrição/complicações , Exposição Materna/efeitos adversos , Metilação , Obesidade/complicações , Obesidade/fisiopatologia , Peptidil Dipeptidase A/genética , Peptidil Dipeptidase A/metabolismo , Gravidez , Complicações na Gravidez/fisiopatologia , Processamento de Proteína Pós-Traducional , Receptor Tipo 1 de Angiotensina/genética , Receptor Tipo 1 de Angiotensina/metabolismo , Sistema Renina-Angiotensina , Carneiro Doméstico , Austrália do Sul , Redução de Peso
20.
Clin Exp Pharmacol Physiol ; 40(11): 803-16, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-24033542

RESUMO

With the worldwide obesity epidemic, the proportion of women entering pregnancy overweight or obese has increased significantly in recent years. Babies born to obese women are at an increased risk of respiratory complications at birth and in childhood. In addition to maternal diabetes, there are a number of metabolic changes that the fetus of an overnourished mother experiences in utero that may modulate lung development and represent the mechanisms underlying the increased risk of respiratory complications. Herein we highlight a series of factors associated with the intrauterine environment of an overnourished mother that may impact on fetal lung development and lead to an increased risk of complications at birth or in postnatal life.


Assuntos
Modelos Animais de Doenças , Desenvolvimento Fetal , Pulmão/embriologia , Fenômenos Fisiológicos da Nutrição Materna , Hipernutrição/fisiopatologia , Animais , Secreções Corporais/metabolismo , Criança , Desenvolvimento Infantil , Pré-Escolar , Diabetes Gestacional/epidemiologia , Diabetes Gestacional/etiologia , Feminino , Humanos , Lactente , Recém-Nascido , Pulmão/crescimento & desenvolvimento , Pulmão/metabolismo , Pulmão/patologia , Masculino , Obesidade/patologia , Obesidade/fisiopatologia , Hipernutrição/patologia , Gravidez , Alvéolos Pulmonares/crescimento & desenvolvimento , Alvéolos Pulmonares/metabolismo , Alvéolos Pulmonares/patologia , Transtornos Respiratórios/epidemiologia , Transtornos Respiratórios/etiologia , Risco
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA