Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Cell Mol Life Sci ; 81(1): 151, 2024 Mar 25.
Artigo em Inglês | MEDLINE | ID: mdl-38526599

RESUMO

Obesity and gestational diabetes (GDM) impact fetal growth during pregnancy. Iron is an essential micronutrient needed for energy-intense feto-placental development, but if mis-handled can lead to oxidative stress and ferroptosis (iron-dependent cell death). In a mouse model showing maternal obesity and glucose intolerance, we investigated the association of materno-fetal iron handling and placental ferroptosis, oxidative damage and stress signalling activation with fetal growth. Female mice were fed a standard chow or high fat, high sugar (HFHS) diet during pregnancy and outcomes were measured at day (d)16 or d19 of pregnancy. In HFHS-fed mice, maternal hepcidin was reduced and iron status maintained (tissue iron levels) at both d16 and d19. However, fetal weight, placental iron transfer capacity, iron deposition, TFR1 expression and ERK2-mediated signalling were reduced and oxidative damage-related lipofuscin accumulation in the placenta was increased in HFHS-fed mice. At d19, whilst TFR1 remained decreased, fetal weight was normal and placental weight, iron content and iron transporter genes (Dmt1, Zip14, and Fpn1) were reduced in HFHS-fed mice. Furthermore, there was stress kinase activation (increased phosphorylated p38MAPK, total ERK and JNK) in the placenta from HFHS-fed mice at d19. In summary, a maternal HFHS diet during pregnancy impacts fetal growth trajectory in association with changes in placental iron handling, ferroptosis and stress signalling. Downregulation of placental iron transporters in HFHS mice may protect the fetus from excessive oxidative iron. These findings suggest a role for alterations in placental iron homeostasis in determining perinatal outcomes of pregnancies associated with GDM and/or maternal obesity.


Assuntos
Ferroptose , Obesidade Materna , Humanos , Gravidez , Feminino , Animais , Camundongos , Ferro , Peso Fetal , Placenta , Feto , Dieta Hiperlipídica/efeitos adversos
2.
Nutrients ; 15(23)2023 Nov 29.
Artigo em Inglês | MEDLINE | ID: mdl-38068814

RESUMO

INTRODUCTION: Docosahexaenoic acid (DHA) is an n-3 long chain polyunsaturated fatty acid critical for fetal brain development that is transported to the fetus from the mother by the placenta. The lysophosphatidylcholine (LPC) transporter, Major Facilitator Superfamily Domain Containing 2a (MFSD2a), is localized in the basal plasma membrane of the syncytiotrophoblast of the human placenta, and MFSD2a expression correlates with umbilical cord blood LPC-DHA levels in human pregnancy. We hypothesized that placenta-specific knockdown of MFSD2a in pregnant mice reduces phospholipid DHA accumulation in the fetal brain. METHODS: Mouse blastocysts (E3.5) were transduced with an EGFP-expressing lentivirus containing either an shRNA targeting MFSD2a or a non-coding sequence (SCR), then transferred to pseudopregnant females. At E18.5, fetuses were weighed and their placenta, brain, liver and plasma were collected. MFSD2a mRNA expression was determined by qPCR in the brain, liver and placenta and phospholipid DHA was quantified by LC-MS/MS. RESULTS: MFSD2a-targeting shRNA reduced placental mRNA MFSD2a expression by 38% at E18.5 (n = 45, p < 0.008) compared with SCR controls. MFSD2a expression in the fetal brain and liver were unchanged. Fetal brain weight was reduced by 13% (p = 0.006). Body weight, placenta and liver weights were unaffected. Fetal brain phosphatidyl choline and phosphatidyl ethanolamine DHA content was lower in fetuses with placenta-specific MFSD2a knockdown. CONCLUSIONS: Placenta-specific reduction in expression of the LPC-DHA transporter MFSD2a resulted in reduced fetal brain weight and lower phospholipid DHA content in the fetal brain. These data provide mechanistic evidence that placental MFSD2a mediates maternal-fetal transfer of LPC-DHA, which is critical for brain growth.


Assuntos
Ácidos Graxos Ômega-3 , Simportadores , Feminino , Animais , Gravidez , Humanos , Camundongos , Ácidos Docosa-Hexaenoicos , Fosfolipídeos , Cromatografia Líquida , Simportadores/metabolismo , Placenta/metabolismo , Espectrometria de Massas em Tandem , Encéfalo/metabolismo , Proteínas de Membrana Transportadoras/metabolismo , Ácidos Graxos Ômega-3/metabolismo , RNA Interferente Pequeno/metabolismo , RNA Mensageiro/metabolismo
3.
Nutrients ; 14(11)2022 May 30.
Artigo em Inglês | MEDLINE | ID: mdl-35684104

RESUMO

Glucocorticoids have an important role in development of the metabolic phenotype in utero. They act as environmental and maturational signals in adapting feto-placental metabolism to maximize the chances of survival both before and at birth. They influence placental nutrient handling and fetal metabolic processes to support fetal growth, fuel storage and energy production with respect to nutrient availability. More specifically, they regulate the transport, utilization and production of a range of nutrients by the feto-placental tissues that enables greater metabolic flexibility in utero while minimizing any further drain on maternal resources during periods of stress. Near term, the natural rise in fetal glucocorticoid concentrations also stimulates key metabolic adaptations that prepare tissues for the new energy demanding functions after birth. Glucocorticoids, therefore, have a central role in the metabolic communication between the mother, placenta and fetus that optimizes offspring metabolic phenotype for survival to reproductive age. This review discusses the effects of maternal and fetal glucocorticoids on the supply and utilization of nutrients by the feto-placental tissues with particular emphasis on studies using quantitative methods to assess metabolism in rodents and sheep in vivo during late pregnancy. It considers the routes of glucocorticoid overexposure in utero, including experimental administration of synthetic glucocorticoids, and the mechanisms by which these hormones control feto-placental metabolism at the molecular, cellular and systems levels. It also briefly examines the consequences of intrauterine glucocorticoid overexposure for postnatal metabolic health and the generational inheritance of metabolic phenotype.


Assuntos
Glucocorticoides , Placenta , Animais , Feminino , Desenvolvimento Fetal , Feto/metabolismo , Glucocorticoides/metabolismo , Parto , Placenta/metabolismo , Gravidez , Ovinos
4.
J Physiol ; 600(13): 3169-3191, 2022 07.
Artigo em Inglês | MEDLINE | ID: mdl-35545608

RESUMO

Obesity in pregnant women causes fetal cardiac dysfunction and increases offspring cardiovascular disease risk, but its effect on myocardial metabolism is unknown. We hypothesized that maternal obesity alters fetal cardiac expression of metabolism-related genes and shifts offspring myocardial substrate preference from glucose towards lipids. Female mice were fed control or obesogenic diets before and during pregnancy. Fetal hearts were studied in late gestation (embryonic day (E) 18.5; term ≈ E21), and offspring were studied at 3, 6, 9 or 24 months postnatally. Maternal obesity increased heart weight and peroxisome proliferator activated receptor gamma (Pparg) expression in female and male fetuses and caused left ventricular diastolic dysfunction in the adult offspring. Cardiac dysfunction worsened progressively with age in female, but not male, offspring of obese dams, in comparison to age-matched control animals. In 6-month-old offspring, exposure to maternal obesity increased cardiac palmitoyl carnitine-supported mitochondrial respiration in males and reduced myocardial 18 F-fluorodeoxyglucose uptake in females. Cardiac Pparg expression remained higher in adult offspring of obese dams than control dams and was correlated with contractile and metabolic function. Maternal obesity did not affect cardiac palmitoyl carnitine respiration in females or 18 F-fluorodeoxyglucose uptake in males and did not alter cardiac 3 H-oleic acid uptake, pyruvate respiration, lipid content or fatty acid/glucose transporter abundance in offspring of either sex. The results support our hypothesis and show that maternal obesity affects offspring cardiac metabolism in a sex-dependent manner. Persistent upregulation of Pparg expression in response to overnutrition in utero might underpin programmed cardiac impairments mechanistically and contribute to cardiovascular disease risk in children of women with obesity. KEY POINTS: Obesity in pregnant women causes cardiac dysfunction in the fetus and increases lifelong cardiovascular disease risk in the offspring. In this study, we showed that maternal obesity in mice induces hypertrophy of the fetal heart in association with altered expression of genes related to nutrient metabolism. Maternal obesity also alters cardiac metabolism of carbohydrates and lipids in the adult offspring. The results suggest that overnutrition in utero might contribute to increased cardiovascular disease risk in children of women with obesity.


Assuntos
Doenças Cardiovasculares , Cardiopatias , Obesidade Materna , Hipernutrição , Efeitos Tardios da Exposição Pré-Natal , Filhos Adultos , Animais , Cardiomegalia/etiologia , Carnitina , Feminino , Coração Fetal , Humanos , Lipídeos , Masculino , Camundongos , Obesidade/metabolismo , Obesidade Materna/complicações , PPAR gama/genética , Gravidez
5.
Sci Rep ; 12(1): 8611, 2022 05 21.
Artigo em Inglês | MEDLINE | ID: mdl-35597818

RESUMO

Bees are important pollinators in wild and agricultural ecosystems, and understanding the factors driving their global declines is key to maintaining these pollination services. Learning, which has been a focus of previous ecotoxicological studies in bees, may play a key role in driving colony fitness. Here we move beyond the standard single-stressor approach to ask how multiple stressors, an agrochemical (sulfoxaflor, a relatively new insecticide) and a parasite (Crithidia bombi, a prevalent gut parasite of bumblebees), impact learning in the bumblebee Bombus terrestris. We developed a modified version of the classic proboscis extension reflex assay to assess the combined effects of acute oral sulfoxaflor exposure and infection by C. bombi on olfactory learning of bumblebee workers. We found no evidence that either sulfoxaflor, C. bombi, or their combination had any significant effect on bumblebee olfactory learning, despite their known negative impacts on other aspects of bumblebee health. This suggests that losses in cognitive ability, as measured here, are unlikely to explain the impacts of sulfoxaflor and its interactions with other stressors on bumblebees. Our novel methodology provides a model system within which to test interactive effects of other key stressors on bee health.


Assuntos
Parasitos , Trypanosoma , Animais , Abelhas , Crithidia , Ecossistema , Piridinas , Compostos de Enxofre
6.
Clin Sci (Lond) ; 135(17): 2049-2066, 2021 09 17.
Artigo em Inglês | MEDLINE | ID: mdl-34406367

RESUMO

Fetal growth restriction (FGR) is a complication of pregnancy that reduces birth weight, markedly increases infant mortality and morbidity and is associated with later-life cardiometabolic disease. No specific treatment is available for FGR. Placentas of human FGR infants have low abundance of sodium-coupled neutral amino acid transporter 2 (Slc38a2/SNAT2), which supplies the fetus with amino acids required for growth. We determined the mechanistic role of placental Slc38a2/SNAT2 deficiency in the development of restricted fetal growth, hypothesizing that placenta-specific Slc38a2 knockdown causes FGR in mice. Using lentiviral transduction of blastocysts with a small hairpin RNA (shRNA), we achieved 59% knockdown of placental Slc38a2, without altering fetal Slc38a2 expression. Placenta-specific Slc38a2 knockdown reduced near-term fetal and placental weight, fetal viability, trophoblast plasma membrane (TPM) SNAT2 protein abundance, and both absolute and weight-specific placental uptake of the amino acid transport System A tracer, 14C-methylaminoisobutyric acid (MeAIB). We also measured human placental SLC38A2 gene expression in a well-defined term clinical cohort and found that SLC38A2 expression was decreased in late-onset, but not early-onset FGR, compared with appropriate for gestational age (AGA) control placentas. The results demonstrate that low placental Slc38a2/SNAT2 causes FGR and could be a target for clinical therapies for late-onset FGR.


Assuntos
Sistema A de Transporte de Aminoácidos/deficiência , Desenvolvimento Fetal , Retardo do Crescimento Fetal/metabolismo , Placenta/metabolismo , Placentação , Sistema A de Transporte de Aminoácidos/genética , Animais , Estudos de Casos e Controles , Feminino , Retardo do Crescimento Fetal/genética , Retardo do Crescimento Fetal/fisiopatologia , Regulação da Expressão Gênica no Desenvolvimento , Técnicas de Silenciamento de Genes , Placenta/fisiopatologia , Gravidez , Estudos Prospectivos , Interferência de RNA
7.
Int J Obes (Lond) ; 44(2): 488-499, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31076636

RESUMO

BACKGROUND/OBJECTIVES: Adiponectin concentrations are low in obese pregnant women. Restoring normal adiponectin concentrations by infusion in obese pregnant mice prevents placental dysfunction, foetal overgrowth and metabolic syndrome in the offspring. We hypothesised that normalising maternal adiponectin in obese late pregnant dams prevents cardiac dysfunction in the adult offspring. SUBJECTS/METHODS: Pregnant female mice with diet-induced obesity were infused with adiponectin (0.62 µg g-1 day-1, n = 24) or saline (n = 22) over days 14.5-18.5 of pregnancy (term = day 19.5). Control dams ate standard chow and received saline (n = 22). Offspring were studied at 3 and 6 months of age. RESULTS: Maternal obesity impaired ventricular diastolic function, increased cardiomyocyte cross-sectional area and upregulated cardiac brain natriuretic peptide (Nppb) and α-skeletal actin (Acta1) gene expression in adult male offspring, compared to control offspring. In adult female offspring, maternal obesity increased Nppb expression, decreased end-diastolic volume and caused age-dependent diastolic dysfunction but not cardiomyocyte hypertrophy. Maternal obesity also activated cardiac Akt and mechanistic target of rapamycin (mTOR) signalling in male, but not in female, offspring and inhibited cardiac extracellular signal-regulated kinase 1/2 (ERK1/2) in both sexes. Normalising maternal circulating adiponectin concentrations by infusing obese dams with adiponectin prevented offspring diastolic dysfunction and ventricular dilation and normalised cardiac Akt-mTOR signalling irrespective of sex. Maternal adiponectin infusion also reduced cardiac Nppb expression and increased ERK1/2 signalling in offspring of obese dams. Adiponectin infusion did not prevent cardiomyocyte hypertrophy but reduced ventricular wall thickness in male offspring and increased collagen content in female offspring of obese dams, compared to controls. CONCLUSIONS: Low maternal adiponectin levels in obese mice in late pregnancy are mechanistically linked to in utero programming of cardiac dysfunction in their offspring. Interventions enhancing endogenous adiponectin secretion or signalling in obese pregnant women could prevent the development of cardiac dysfunction in their children.


Assuntos
Adiponectina , Cardiopatias/prevenção & controle , Fenômenos Fisiológicos da Nutrição Materna/efeitos dos fármacos , Efeitos Tardios da Exposição Pré-Natal/prevenção & controle , Adiponectina/administração & dosagem , Adiponectina/sangue , Adiponectina/farmacologia , Animais , Ecocardiografia , Feminino , Coração/diagnóstico por imagem , Coração/fisiopatologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Obesos , Miocárdio/patologia , Gravidez
8.
J Appl Physiol (1985) ; 128(1): 127-133, 2020 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-31804891

RESUMO

Women residing at high altitudes deliver infants of lower birth weight than at sea level. Birth weight correlates with placental system A-mediated amino acid transport capacity, and severe environmental hypoxia reduces system A activity in isolated trophoblast and the mouse placenta. However, the effect of high altitude on human placental amino acid transport remains unknown. We hypothesized that microvillous membrane (MVM) system A and system L amino acid transporter activity is lower in placentas of women living at high altitude compared with low-altitude controls. Placentas were collected at term from healthy pregnant women residing at high altitude (HA; >2,500 m; n = 14) or low altitude (LA; <1,700 m; n = 14) following planned, unlabored cesarean section. Birth weight, but not placenta weight, was 13% lower in HA pregnancies (2.88 ± 0.11 kg) compared with LA (3.30 ± 0.07 kg, P < 0.01). MVM erythropoietin receptor abundance, determined by immunoblot, was greater in HA than in LA placentas, consistent with lower placental oxygen levels at HA. However, there was no effect of altitude on MVM system A or L activity, determined by Na+-dependent [14C]methylaminoisobutyric acid uptake and [3H]leucine uptake, respectively. MVM abundance of glucose transporters (GLUTs) 1 and 4 and basal membrane GLUT4 were also similar in LA and HA placentas. Low birth weights in the neonates of women residing at high altitude are not a consequence of reduced placental amino acid transport capacity. These observations are in general agreement with studies of IUGR babies at low altitude, in which MVM system A activity is downregulated only in growth-restricted babies with significant compromise.NEW & NOTEWORTHY Babies born at high altitude are smaller than at sea level. Birth weight is dependent on growth in utero and, in turn, placental nutrient transport. We determined amino acid transport capacity in placentas collected from women resident at low and high altitude. Altitude did not affect system A amino acid transport across the syncytiotrophoblast microvillous membrane, suggesting that impaired placental amino acid transport does not contribute to reduced birth weight in this high-altitude population.


Assuntos
Altitude , Aminoácidos/metabolismo , Peso ao Nascer , Retardo do Crescimento Fetal/metabolismo , Placenta/metabolismo , Trofoblastos/metabolismo , Adulto , Transporte Biológico , Feminino , Retardo do Crescimento Fetal/patologia , Humanos , Masculino , Placenta/patologia , Gravidez , Trofoblastos/patologia , Adulto Jovem
9.
Proc Natl Acad Sci U S A ; 116(5): 1621-1626, 2019 01 29.
Artigo em Inglês | MEDLINE | ID: mdl-30655345

RESUMO

Mitochondria respond to a range of stimuli and function in energy production and redox homeostasis. However, little is known about the developmental and environmental control of mitochondria in the placenta, an organ vital for fetal growth and pregnancy maintenance in eutherian mammals. Using respirometry and molecular analyses, the present study examined mitochondrial function in the distinct transport and endocrine zones of the mouse placenta during normal pregnancy and maternal inhalation hypoxia. The data show that mitochondria of the two zones adopt different strategies in modulating their respiration, substrate use, biogenesis, density, and efficiency to best support the growth and energy demands of fetoplacental tissues during late gestation in both normal and hypoxic conditions. The findings have important implications for environmentally induced adaptations in mitochondrial function in other tissues and for compromised human pregnancy in which hypoxia and alterations in placental mitochondrial function are associated with poor outcomes like fetal growth restriction.


Assuntos
Desenvolvimento Fetal/fisiologia , Hipóxia/fisiopatologia , Mitocôndrias/fisiologia , Placenta/fisiopatologia , Animais , Feminino , Retardo do Crescimento Fetal/fisiopatologia , Camundongos , Camundongos Endogâmicos C57BL , Gravidez
10.
J Physiol ; 595(14): 4875-4892, 2017 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-28382681

RESUMO

KEY POINTS: In the Western world, obesogenic diets containing high fat and high sugar (HFHS) are commonly consumed during pregnancy, although their effects on the metabolism of the mother, in relation to feto-placental glucose utilization and growth, are unknown. In the present study, the consumption of an obesogenic HFHS diet compromised maternal glucose tolerance and insulin sensitivity in late pregnancy in association with dysregulated lipid and glucose handling by the dam. These maternal metabolic changes induced by HFHS feeding were related to altered feto-placental glucose metabolism and growth. A HFHS diet during pregnancy therefore causes maternal metabolic dysfunction with consequences for maternal nutrient allocation for fetal growth. These findings have implications for the health of women and their infants, who consume obesogenic diets during pregnancy. ABSTRACT: In the Western world, obesogenic diets containing high fat and high sugar (HFHS) are commonly consumed during pregnancy. However, the impacts of a HFHS diet during pregnancy on maternal insulin sensitivity and signalling in relation to feto-placental growth and glucose utilization are unknown. The present study examined the effects of a HFHS diet during mouse pregnancy on maternal glucose tolerance and insulin resistance, as well as, on feto-placental glucose metabolism. Female mice were fed a control or HFHS diet from day (D) 1 of pregnancy (term = D20.5). At D16 or D19, dams were assessed for body composition, metabolite and hormone concentrations, tissue abundance of growth and metabolic signalling pathways, glucose tolerance and utilization and insulin sensitivity. HFHS feeding perturbed maternal insulin sensitivity in late pregnancy; hepatic insulin sensitivity was higher, whereas sensitivity of the skeletal muscle and white adipose tissue was lower in HFHS than control dams. These changes were accompanied by increased adiposity and reduced glucose production and glucose tolerance of HFHS dams. The HFHS diet also disturbed the hormone and metabolite milieu and altered expression of growth and metabolic signalling pathways in maternal tissues. Furthermore, HFHS feeding was associated with impaired feto-placental glucose metabolism and growth. A HFHS diet during pregnancy therefore causes maternal metabolic dysfunction with consequences for maternal nutrient allocation for fetal growth. These findings have implications for the health of women and their infants, who consume HFHS diets during pregnancy.


Assuntos
Dieta Ocidental , Feto/metabolismo , Glucose/metabolismo , Obesidade/metabolismo , Placenta/metabolismo , Tecido Adiposo Branco/metabolismo , Animais , Glicemia/análise , Feminino , Desenvolvimento Fetal , Teste de Tolerância a Glucose , Insulina , Resistência à Insulina , Metabolismo dos Lipídeos , Fígado/metabolismo , Camundongos Endogâmicos C57BL , Mães , Músculo Esquelético/metabolismo , Gravidez
11.
Hum Gene Ther ; 27(12): 997-1007, 2016 12.
Artigo em Inglês | MEDLINE | ID: mdl-27530140

RESUMO

In a model of growth-restricted sheep pregnancy, it was previously demonstrated that transient uterine artery VEGF overexpression can improve fetal growth. This approach was tested in guinea-pig pregnancies, where placental physiology is more similar to humans. Fetal growth restriction (FGR) was attained through peri-conceptual nutrient restriction in virgin guinea pigs. Ad.VEGF-A165 or Ad.LacZ (1 × 1010vp) was applied at mid-gestation via laparotomy, delivered externally to the uterine circulation with thermosensitive gel. At short-term (3-8 days post surgery) or at term gestation, pups were weighed, and tissues were sampled for vector spread analysis, VEGF expression, and its downstream effects. Fetal weight at term was increased (88.01 ± 13.36 g; n = 26) in Ad.VEGF-A165-treated animals compared with Ad.LacZ-treated animals (85.52 ± 13.00 g; n = 19; p = 0.028). The brain, liver, and lung weight and crown rump length were significantly larger in short-term analyses, as well as VEGF expression in transduced tissues. At term, molecular analyses confirmed the presence of VEGF transgene in target tissues but not in fetal samples. Tissue histology analysis and blood biochemistry/hematological examination were comparable with controls. Uterine artery relaxation in Ad.VEGF-A165-treated dams was higher compared with Ad.LacZ-treated dams. Maternal uterine artery Ad.VEGF-A165 increases fetal growth velocity and term fetal weight in growth-restricted guinea-pig pregnancy.


Assuntos
Adenoviridae/genética , Retardo do Crescimento Fetal/genética , Retardo do Crescimento Fetal/terapia , Peso Fetal/genética , Terapia Genética , Vetores Genéticos/administração & dosagem , Fator A de Crescimento do Endotélio Vascular/genética , Animais , Feminino , Cobaias , Gravidez , Fluxo Sanguíneo Regional
12.
Diabetes ; 65(4): 851-60, 2016 04.
Artigo em Inglês | MEDLINE | ID: mdl-26740602

RESUMO

In late pregnancy, maternal insulin resistance occurs to support fetal growth, but little is known about insulin-glucose dynamics close to delivery. This study measured insulin sensitivity in mice in late pregnancy at day 16 (D16) and near term at D19. Nonpregnant (NP) and pregnant mice were assessed for metabolite and hormone concentrations, body composition by DEXA, tissue insulin signaling protein abundance by Western blotting, glucose tolerance and utilization, and insulin sensitivity using acute insulin administration and hyperinsulinemic-euglycemic clamps with [(3)H]glucose infusion. Whole-body insulin resistance occurred in D16 pregnant dams in association with basal hyperinsulinemia, insulin-resistant endogenous glucose production, and downregulation of several proteins in hepatic and skeletal muscle insulin signaling pathways relative to NP and D19 values. Insulin resistance was less pronounced at D19, with restoration of NP insulin concentrations, improved hepatic insulin sensitivity, and increased abundance of hepatic insulin signaling proteins. At D16, insulin resistance at whole-body, tissue, and molecular levels will favor fetal glucose acquisition, while improved D19 hepatic insulin sensitivity will conserve glucose for maternal use in anticipation of lactation. Tissue sensitivity to insulin, therefore, alters differentially with proximity to delivery in pregnant mice, with implications for human and other species.


Assuntos
Glucose/metabolismo , Resistência à Insulina/fisiologia , Parto/metabolismo , Gravidez/metabolismo , Animais , Parto Obstétrico , Feminino , Metabolismo dos Lipídeos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Fatores de Tempo
13.
PLoS One ; 10(8): e0136115, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26287800

RESUMO

The effects of endogenous and synthetic glucocorticoids on fetal lung maturation are well-established, although the role of leptin in lung development before birth is unclear. This study examined mRNA and protein levels of the signalling long-form leptin receptor (Ob-Rb) in fetal ovine lungs towards term, and after experimental manipulation of glucocorticoid levels in utero by fetal cortisol infusion or maternal dexamethasone treatment. In fetal ovine lungs, Ob-Rb protein was localised to bronchiolar epithelium, bronchial cartilage, vascular endothelium, alveolar macrophages and type II pneumocytes. Pulmonary Ob-Rb mRNA abundance increased between 100 (0.69 fractional gestational age) and 144 days (0.99) of gestation, and by 2-4-fold in response to fetal cortisol infusion and maternal dexamethasone treatment. In contrast, pulmonary Ob-Rb protein levels decreased near term and were halved by glucocorticoid treatment, without any significant change in phosphorylated signal transducer and activator of transcription-3 (pSTAT3) at Ser727, total STAT3 or the pulmonary pSTAT3:STAT3 ratio. Leptin mRNA was undetectable in fetal ovine lungs at the gestational ages studied. These findings demonstrate differential control of pulmonary Ob-Rb transcript abundance and protein translation, and/or post-translational processing, by glucocorticoids in utero. Localisation of Ob-Rb in the fetal ovine lungs, including alveolar type II pneumocytes, suggests a role for leptin signalling in the control of lung growth and maturation before birth.


Assuntos
Pulmão/embriologia , Pulmão/metabolismo , Receptores para Leptina/genética , Receptores para Leptina/metabolismo , Células Epiteliais Alveolares/metabolismo , Animais , Dexametasona/farmacologia , Feminino , Feto/metabolismo , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Glucocorticoides/farmacologia , Hidrocortisona/sangue , Hidrocortisona/farmacologia , Pulmão/efeitos dos fármacos , Fosforilação , Gravidez , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Fator de Transcrição STAT3/química , Fator de Transcrição STAT3/metabolismo , Carneiro Doméstico , Transdução de Sinais
14.
Biol Reprod ; 89(4): 80, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23986571

RESUMO

Synthetic glucocorticoids, like dexamethasone (dex), restrict growth of the fetus and program its adult physiology, in part by altering placental phenotype. The route and timing of dex administration determine the fetal and adult outcomes, but whether these factors affect placental phenotype remains unknown. This study compared placental morphology, amino acid transport, and gene expression in mice given dex orally or by subcutaneous injection over the periods of most rapid placental (Days [D] 11-16) or fetal (D14-19) growth (term is D21). Compared with untreated and saline-injected controls, both dex treatments reduced placental weight at D16 and 19 and fetal weight and total labyrinthine volume at D19 to a similar extent. Only oral dex treatment from D11 to D16 reduced labyrinthine fetal capillary volume on D16 and increased placental ¹4C-methylaminoisobutyric acid (MeAIB) clearance at D19, 3 days after treatment ended. Neither route of dex treatment altered placental expression of Slc38a, Hsd11b, or the glucocorticoid receptor, Nr3c1, at D16. In contrast, both routes of dex treatment from D14 to D19 increased placental Hsd11b2 expression and labyrinthine maternal vessel volume. Furthermore, injection per se altered placental expression of Nr3c1, Hsd11b1, and specific Slc38a isoforms in an age-related manner. Overall, MeAIB clearance was not related to Slc38a transporter expression but was correlated inversely with maternal corticosterone concentrations when dex was undetectable in maternal plasma at D19. The effects of dex on placental phenotype, therefore, depend on both the route and timing of administration and may relate to local glucocorticoid availability during and after the treatment period.


Assuntos
Dexametasona/administração & dosagem , Desenvolvimento Fetal/efeitos dos fármacos , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Glucocorticoides/administração & dosagem , Troca Materno-Fetal/efeitos dos fármacos , Placenta/efeitos dos fármacos , Placentação/efeitos dos fármacos , Administração Oral , Aminoácidos/metabolismo , Animais , Transporte Biológico/efeitos dos fármacos , Corticosterona/sangue , Dexametasona/efeitos adversos , Dexametasona/sangue , Dexametasona/farmacocinética , Feminino , Retardo do Crescimento Fetal/induzido quimicamente , Retardo do Crescimento Fetal/metabolismo , Retardo do Crescimento Fetal/patologia , Peso Fetal/efeitos dos fármacos , Glucocorticoides/efeitos adversos , Glucocorticoides/sangue , Glucocorticoides/farmacocinética , Injeções Subcutâneas , Camundongos , Camundongos Endogâmicos C57BL , Placenta/irrigação sanguínea , Placenta/metabolismo , Circulação Placentária/efeitos dos fármacos , Gravidez , Proteínas da Gravidez/genética , Proteínas da Gravidez/metabolismo , Distribuição Aleatória
15.
FASEB J ; 27(10): 3928-37, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23825226

RESUMO

In developed societies, high-sugar and high-fat (HSHF) diets are now the norm and are increasing the rates of maternal obesity during pregnancy. In pregnant rodents, these diets lead to cardiovascular and metabolic dysfunction in their adult offspring, but the intrauterine mechanisms involved remain unknown. This study shows that, relative to standard chow, HSHF feeding throughout mouse pregnancy increases maternal adiposity (+30%, P<0.05) and reduces fetoplacental growth at d 16 (-10%, P<0.001). At d 19, however, HSHF diet group pup weight had normalized, despite the HSHF diet group placenta remaining small and morphologically compromised. This altered fetal growth trajectory was associated with enhanced placental glucose and amino acid transfer (+35%, P<0.001) and expression of their transporters (+40%, P<0.024). HSHF feeding also up-regulated placental expression of fatty acid transporter protein, metabolic signaling pathways (phosphoinositol 3-kinase and mitogen-activated protein kinase), and several growth regulatory imprinted genes (Igf2, Dlk1, Snrpn, Grb10, and H19) independently of changes in DNA methylation. Obesogenic diets during pregnancy, therefore, alter maternal nutrient partitioning, partly through changes in the placental phenotype, which helps to meet fetal nutrient demands for growth near term. However, by altering provision of specific nutrients, dietary-induced placental adaptations have important roles in programming development with health implications for the offspring in later life.


Assuntos
Gorduras na Dieta/farmacologia , Desenvolvimento Fetal/efeitos dos fármacos , Fenômenos Fisiológicos da Nutrição Materna , Fenômenos Fisiológicos da Nutrição Animal , Animais , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , Placenta/metabolismo , Gravidez
16.
Curr Opin Clin Nutr Metab Care ; 16(3): 298-309, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23340010

RESUMO

PURPOSE OF REVIEW: Size at birth is critical in determining life expectancy with both small and large neonates at risk of shortened life spans. This review examines the hormonal and nutritional drivers of intrauterine growth with emphasis on the role of foetal hormones as nutritional signals in utero. RECENT FINDINGS: Nutrients drive intrauterine growth by providing substrate for tissue accretion, whereas hormones regulate nutrient distribution between foetal oxidative metabolism and mass accumulation. The main hormonal drivers of intrauterine growth are insulin, insulin-like growth factors and thyroid hormones. Together with leptin and cortisol, these hormones control cellular nutrient uptake and the balance between accretion and differentiation in regulating tissue growth. They also act indirectly via the placenta to alter the materno-foetal supply of nutrients and oxygen. By responding to nutrient and oxygen availability, foetal hormones optimize the survival and growth of the foetus with respect to its genetic potential, particularly during adverse conditions. However, changes in the intrauterine growth of individual tissues may alter their function permanently. SUMMARY: In both normal and compromised pregnancies, intrauterine growth is determined by multiple hormonal and nutritional drivers which interact to produce a specific pattern of intrauterine development with potential lifelong consequences for health.


Assuntos
Desenvolvimento Fetal , Feto/metabolismo , Fenômenos Fisiológicos da Nutrição Materna , Estado Nutricional , Hormônios Tireóideos/metabolismo , Disponibilidade Biológica , Feminino , Feto/embriologia , Humanos , Insulina/metabolismo , Leptina/metabolismo , Oxigênio/administração & dosagem , Oxigênio/farmacocinética , Placenta/embriologia , Placenta/metabolismo , Placentação , Gravidez , Somatomedinas/metabolismo
17.
J Physiol ; 590(21): 5529-40, 2012 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-22930269

RESUMO

Stresses during pregnancy that increase maternal glucocorticoids reduce birth weight in several species. However, the role of natural glucocorticoids in the mother in fetal acquisition of nutrients for growth remains unknown. This study aimed to determine whether fetal growth was reduced as a consequence of altered amino acid supply when mice were given corticosterone in their drinking water for 5 day periods in mid to late pregnancy (day, D, 11-16 or D14-19). Compared to controls drinking tap water, fetal weight was always reduced by corticosterone. At D16, corticosterone had no effect on materno-fetal transfer of [(14)C]methylaminoisobutyric acid (MeAIB), although placental MeAIB accumulation and expression of the Slc38a1 and Slc38a2 transporters were increased. However, at D19, 3 days after treatment ended, materno-fetal transfer of MeAIB was increased by 37% (P < 0.04). During treatment at D19, placental accumulation and materno-fetal transfer of MeAIB were reduced by 40% (P < 0.01), although expression of Slc38a1 was again elevated. Permanent reductions in placental vascularity occurred during the earlier but not the later period of treatment. Placental Hsd11b2 expression, which regulates feto-placental glucocorticoid bioavailability, was also affected by treatment at D19 only. Maternal corticosterone concentrations inversely correlated with materno-fetal MeAIB clearance and fetal weight at D19 but not D16. On D19, weight gain of the maternal carcass was normal during corticosterone treatment but reduced in those mice treated from D11 to D16, in which corticosterone levels were lowest. Maternal corticosterone is, therefore, a physiological regulator of the amino acid supply for fetal growth via actions on placental phenotype.


Assuntos
Corticosterona/fisiologia , Desenvolvimento Fetal/fisiologia , Prenhez/fisiologia , 11-beta-Hidroxiesteroide Desidrogenase Tipo 2/genética , Sistema A de Transporte de Aminoácidos/genética , Aminoácidos/metabolismo , Animais , Feminino , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Placenta/metabolismo , Placentação , Gravidez , RNA Mensageiro/metabolismo
18.
Mol Cell ; 11(4): 1043-54, 2003 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-12718889

RESUMO

p300 and CREB binding protein can both activate and repress transcription. Here, we locate the CRD1 transcriptional repression domain between residues 1017 and 1029 of p300. This region contains two copies of the sequence psiKxE that are modified by the ubiquitin-like protein SUMO-1. Mutations that reduce SUMO modification increase p300-mediated transcriptional activity and expression of a SUMO-specific protease or catalytically inactive Ubc9 relieved repression, demonstrating that p300 repression was mediated by SUMO conjugation. SUMO-modified CRD1 domain bound HDAC6 in vitro, and p300 repression was relieved by histone deacetylase inhibition and siRNA-mediated ablation of HDAC6 expression. These results reveal a mechanism controlling p300 function and suggest that SUMO-dependent repression is mediated by recruitment of HDAC6.


Assuntos
Células Eucarióticas/metabolismo , Genes Reguladores/genética , Proteínas Nucleares/metabolismo , Proteínas Repressoras/metabolismo , Proteína SUMO-1/metabolismo , Transativadores/metabolismo , Enzimas de Conjugação de Ubiquitina , Sequência de Aminoácidos/genética , Sítios de Ligação/genética , Estruturas do Núcleo Celular/genética , Estruturas do Núcleo Celular/metabolismo , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Inibidores Enzimáticos/farmacologia , Células HeLa , Desacetilase 6 de Histona , Inibidores de Histona Desacetilases , Histona Desacetilases/genética , Histona Desacetilases/metabolismo , Humanos , Ligases/genética , Ligases/metabolismo , Mutação/genética , Proteínas Nucleares/genética , Estrutura Terciária de Proteína/genética , RNA Interferente Pequeno , Proteínas Repressoras/genética , Proteína SUMO-1/genética , Transativadores/genética , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...