Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
1.
J Biochem Mol Toxicol ; 35(1): e22629, 2021 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-32935389

RESUMO

Gestational diabetes mellitus is one of the causes of abnormal embryonic heart development, but the mechanism is still poor. This study investigated the regulatory mechanism and role of SOX11 in congenital heart abnormality in a hyperglycemic environment. Immunohistochemistry, Western blotting, and quantitative reverse transcription-polymerase chain reaction (qRT-PCR) showed decreased SOX11 protein and messenger RNA (mRNA) levels in the heart tissue of diabetic offspring compared with the control group. A Sequenom EpiTYPER MassArray showed that methylation sites upstream in SOX11 region 1 were increased in the diabetic group compared with the control group. Luciferase reporter assays and qRT-PCR showed that Dnmt3b overexpression decreased SOX11 promoter activity and its mRNA level, whereas Dnmt3a had little effect on regulating SOX11 expression. Furthermore, we found that Dnmt3L cooperated with Dnmt3b to regulate SOX11 gene expression. Additionally, the function of SOX11 silencing was analyzed by using small interfering RNA-mediated knockdown. 3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide and apoptotic assays showed that SOX11 downregulation inhibited cell viability and induced apoptosis in cardiomyocytes. Overexpression of the SOX11 gene suppressed cardiomyocytes apoptosis after high glucose treatment. We identified a novel epigenetic regulatory mechanism of SOX11 during heart development in a hyperglycemic environment and revealed a distinct role of SOX11 in mediating cardiomyocytes viability and apoptosis.


Assuntos
Apoptose , Regulação para Baixo , Feto/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Hiperglicemia/embriologia , Miocárdio/metabolismo , Miócitos Cardíacos/metabolismo , Fatores de Transcrição SOXC/biossíntese , Animais , Feminino , Feto/patologia , Hiperglicemia/patologia , Masculino , Miocárdio/patologia , Miócitos Cardíacos/patologia , Ratos , Ratos Sprague-Dawley
2.
Proc Natl Acad Sci U S A ; 115(40): 10142-10147, 2018 10 02.
Artigo em Inglês | MEDLINE | ID: mdl-30224493

RESUMO

The primary stem cells of the cerebral cortex are the radial glial cells (RGCs), and disturbances in their operation lead to myriad brain disorders in all mammals from mice to humans. Here, we found in mice that maternal gestational obesity and hyperglycemia can impair the maturation of RGC fibers and delay cortical neurogenesis. To investigate potential mechanisms, we used optogenetic live-imaging approaches in embryonic cortical slices. We found that Ca2+ signaling regulates mitochondrial transport and is crucial for metabolic support in RGC fibers. Cyclic intracellular Ca2+ discharge from localized RGC fiber segments detains passing mitochondria and ensures their proper distribution and enrichment at specific sites such as endfeet. Impairment of mitochondrial function caused an acute loss of Ca2+ signaling, while hyperglycemia decreased Ca2+ activity and impaired mitochondrial transport, leading to degradation of the RGC scaffold. Our findings uncover a physiological mechanism indicating pathways by which gestational metabolic disturbances can interfere with brain development.


Assuntos
Sinalização do Cálcio , Córtex Cerebral/embriologia , Diabetes Gestacional/metabolismo , Glucose/metabolismo , Hiperglicemia/embriologia , Neurogênese , Neuroglia/metabolismo , Animais , Córtex Cerebral/patologia , Diabetes Gestacional/genética , Diabetes Gestacional/patologia , Feminino , Hiperglicemia/genética , Hiperglicemia/patologia , Camundongos , Camundongos Transgênicos , Mitocôndrias/genética , Mitocôndrias/metabolismo , Mitocôndrias/patologia , Neuroglia/patologia , Gravidez
3.
Cell Tissue Res ; 368(3): 563-578, 2017 06.
Artigo em Inglês | MEDLINE | ID: mdl-28283910

RESUMO

Exposure to maternal diabetes during fetal growth is a risk factor for the development of type II diabetes (T2D) in later life. Discovery of the mechanisms involved in this association should provide valuable background for therapeutic treatments. Early embryogenesis involves epigenetic changes including histone modifications. The bivalent histone methylation marks H3K4me3 and H3K27me3 are important for regulating key developmental genes during early fetal pancreas specification. We hypothesized that maternal hyperglycemia disrupted early pancreas development through changes in histone bivalency. A human embryonic stem cell line (VAL3) was used as the cell model for studying the effects of hyperglycemia upon differentiation into definitive endoderm (DE), an early stage of the pancreatic lineage. Hyperglycemic conditions significantly down-regulated the expression levels of DE markers SOX17, FOXA2, CXCR4 and EOMES during differentiation. This was associated with retention of the repressive histone methylation mark H3K27me3 on their promoters under hyperglycemic conditions. The disruption of histone methylation patterns was observed as early as the mesendoderm stage, with Wnt/ß-catenin signaling being suppressed during hyperglycemia. Treatment with Wnt/ß-catenin signaling activator CHIR-99021 restored the expression levels and chromatin methylation status of DE markers, even in a hyperglycemic environment. The disruption of DE development was also found in mouse embryos at day 7.5 post coitum from diabetic mothers. Furthermore, disruption of DE differentiation in VAL3 cells led to subsequent impairment in pancreatic progenitor formation. Thus, early exposure to hyperglycemic conditions hinders DE development with a possible relationship to the later impairment of pancreas specification.


Assuntos
Diferenciação Celular , Endoderma/citologia , Histonas/metabolismo , Hiperglicemia/embriologia , Pâncreas/embriologia , Animais , Antígenos de Diferenciação/genética , Antígenos de Diferenciação/metabolismo , Azacitidina/farmacologia , Linhagem Celular , Linhagem da Célula , Metilases de Modificação do DNA/antagonistas & inibidores , Metilases de Modificação do DNA/metabolismo , Endoderma/metabolismo , Proteína Potenciadora do Homólogo 2 de Zeste/metabolismo , Inibidores Enzimáticos/farmacologia , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Glucose/farmacologia , Humanos , Hiperglicemia/metabolismo , Masculino , Mesoderma/metabolismo , Metilação , Camundongos , Camundongos Endogâmicos ICR , Pâncreas/citologia , Pâncreas/metabolismo , Regiões Promotoras Genéticas , Transdução de Sinais , Proteínas Wnt/metabolismo , beta Catenina/metabolismo
4.
Am J Obstet Gynecol ; 212(5): 650.e1-11, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25595579

RESUMO

OBJECTIVE: Oxidative stress plays a causal role in diabetic embryopathy. Maternal diabetes induces heart defects and impaired transforming growth factor beta (TGFß) signaling, which is essential for cardiogenesis. We hypothesize that mitigating oxidative stress through superoxide dismutase 1 (SOD1) overexpression in transgenic (Tg) mice reverses maternal hyperglycemia-impaired TGFß signaling and its downstream effectors. STUDY DESIGN: Day 12.5 embryonic hearts from wild-type (WT) and SOD1 overexpressing embryos of nondiabetic (ND) and diabetic mellitus (DM) dams were used for the detection of oxidative stress markers: 4-hydroxynonenal (4-HNE) and malondlaldehyde (MDA), and TGFß1, 2, and 3, phosphor (p)-TGFß receptor II (TßRII), p-phosphorylated mothers against decapentaplegic (Smad)2, and p-Smad3. The expression of 3 TGFß-responsive genes was also assessed. Day 11.5 embryonic hearts were explanted and cultured ex vivo, with or without treatments of a SOD1 mimetic (Tempol; Enzo Life Science, Farmingdale, NY) or a TGFß recombinant protein for the detection of TGFß signaling intermediates. RESULTS: Levels of 4-HNE and MDA were significantly increased by maternal diabetes, and SOD1 overexpression blocked the increase of these 2 oxidative stress markers. Maternal diabetes suppresses the TGFß signaling pathway by down-regulating TGFß1 and TGFß3 expression. Consequently, phosphorylation of TßRII, Smad2, and Smad3, downstream effectors of TGFß, and expression of 3 TGFß-responsive genes were reduced by maternal diabetes, and these reductions were prevented by SOD1 overexpression. Treatment with Tempol or TGFß recombinant protein restored high-glucose-suppressed TGFß signaling intermediates and responsive gene expression. CONCLUSION: Oxidative stress mediates the inhibitory effect of hyperglycemia in the developing heart. Antioxidants, TGFß recombinant proteins, or TGFß agonists may have potential therapeutic values in the prevention of heart defects in diabetic pregnancies.


Assuntos
Coração/embriologia , Hiperglicemia/metabolismo , Miocárdio/metabolismo , Estresse Oxidativo , Gravidez em Diabéticas/metabolismo , Fator de Crescimento Transformador beta1/metabolismo , Fator de Crescimento Transformador beta2/metabolismo , Fator de Crescimento Transformador beta3/metabolismo , Aldeídos/metabolismo , Animais , Feminino , Hiperglicemia/embriologia , Malondialdeído/metabolismo , Camundongos , Camundongos Transgênicos , Fosforilação , Gravidez , Proteínas Serina-Treonina Quinases/metabolismo , Receptor do Fator de Crescimento Transformador beta Tipo II , Receptores de Fatores de Crescimento Transformadores beta/metabolismo , Transdução de Sinais , Proteína Smad2/metabolismo , Proteína Smad3/metabolismo , Superóxido Dismutase/genética , Superóxido Dismutase-1
5.
Am J Obstet Gynecol ; 212(5): 569-79, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25434839

RESUMO

Maternal diabetes-induced birth defects occur in 6-10% of babies born to mothers with pregestational diabetes, representing a significant maternal-fetal health problem. Currently, these congenital malformations represent a significant maternal-fetal medicine issue, but are likely to create an even greater public health threat as 3 million women of reproductive age (19-44 years) have diabetes in the United States alone, and this number is expected to double by 2030. Neural tube defects (NTDs) and congenital heart defects are the most common types of birth defects associated with maternal diabetes. Animal studies have revealed that embryos under hyperglycemic conditions exhibit high levels of oxidative stress resulting from enhanced production of reactive oxygen species and impaired antioxidant capability. Oxidative stress activates a set of proapoptotic kinase signaling intermediates leading to abnormal cell death in the embryonic neural tube, which causes NTD formation. Work in animal models also has revealed that maternal diabetes triggers a series of signaling intermediates: protein kinase C (PKC) isoforms, PKCα, ßII and δ; apoptosis signal-regulating kinase 1; c-Jun-N-terminal kinase (JNK)1/2; caspase; and apoptosis. Specifically, maternal diabetes in rodent models activates the proapoptotic unfolded protein response and endoplasmic reticulum (ER) stress. A reciprocal causation between JNK1/2 activation and ER stress exists in diabetic embryopathy. Molecular studies further demonstrate that deletion of the genes for Prkc, Ask1, Jnk1, or Jnk2 abolishes maternal diabetes-induced neural progenitor apoptosis and ameliorates NTD formation. Similar preventive effects are also observed when apoptosis signal-regulating kinase 1, JNK1/2, or ER stress is inhibited. Cell membrane stabilizers and antioxidant supplements are also effective in prevention of diabetes-induced birth defects. Mechanistic studies have revealed important insights into our understanding the cause of diabetic embryopathy and have provided a basis for future interventions against birth defects or other pregnancy complications associated with maternal diabetes. The knowledge of a molecular pathway map identified in animal studies has created unique opportunities to identify molecular targets for therapeutic intervention.


Assuntos
Apoptose , Cardiopatias Congênitas/metabolismo , Hiperglicemia/metabolismo , Defeitos do Tubo Neural/metabolismo , Estresse Oxidativo , Gravidez em Diabéticas/metabolismo , Proteínas Quinases , Estresse do Retículo Endoplasmático , Feminino , Cardiopatias Congênitas/embriologia , Humanos , Hiperglicemia/embriologia , Defeitos do Tubo Neural/embriologia , Gravidez , Transdução de Sinais
6.
Reprod Biol Endocrinol ; 12: 80, 2014 Aug 18.
Artigo em Inglês | MEDLINE | ID: mdl-25135621

RESUMO

BACKGROUND: Fetal adaptations to high fat (HF) diet in utero (IU) that may predispose to Metabolic Syndrome (MetS) in adulthood include changes in fetal hepatic gene expression. Studies were performed to determine whether maternal exposure to HF diet at different stages during pregnancy had different effects on the fetus, including hepatic gene expression. METHODS: Female wild type mice were fed either a HF or breeding chow (C) for 2 wks prior to mating. The experimental groups were composed of embryonic day (e) 18.5 fetuses obtained from WT female mice that were fed HF (HF, 35.5% fat) or breeding chow (C, 9.5% fat) for 2 wk before mating until e9.5 of pregnancy (periconception-midpregnancy). At e9.5 dams were switched to the opposite diet (C-HF or HF-C). RESULTS: Exposure to HF diet throughout pregnancy reduced maternal weight gain compared to C diet (p < 0.02 HF vs. C). HF-C dams had significantly decreased adiponectin levels and litter size when compared to C-HF (p < 0.02 HF-C vs C-HF). Independent of the timing of exposure to HF, fetal weight and length were significantly decreased when compared to C diet (HF, C-HF and HF-C vs. C p < 0.02). HF diet during the second half of pregnancy increased expression of genes in the fetal liver associated with fetal growth (C-HF vs C p < 0.001), glucose production (C-HF vs C p < 0.04), oxidative stress and inflammation (C-HF vs C p < 0.01) compared to C diet. CONCLUSIONS: This model defines that there are critical periods during gestation in which the fetus is actively shaped by the environment. Early exposure to a HF diet determines litter size while exposure to HF during the second half of pregnancy leads to dysregulation of expression of key genes responsible for fetal growth, hepatic glucose production and oxidative stress. These findings underscore the importance of future studies designed to clarify how these critical periods may influence future risk of developing MetS later in life.


Assuntos
Dieta Hiperlipídica/efeitos adversos , Desenvolvimento Fetal , Retardo do Crescimento Fetal/etiologia , Hiperglicemia/etiologia , Fenômenos Fisiológicos da Nutrição Materna , Síndrome Metabólica/etiologia , Estresse Oxidativo , Adiponectina/sangue , Animais , Animais não Endogâmicos , Cruzamentos Genéticos , Feminino , Retardo do Crescimento Fetal/imunologia , Retardo do Crescimento Fetal/metabolismo , Peso Fetal , Regulação da Expressão Gênica no Desenvolvimento , Gluconeogênese , Transportador de Glucose Tipo 4/genética , Hiperglicemia/embriologia , Hiperglicemia/imunologia , Hiperglicemia/metabolismo , Tamanho da Ninhada de Vivíparos , Fígado/embriologia , Fígado/imunologia , Fígado/metabolismo , Síndrome Metabólica/embriologia , Síndrome Metabólica/imunologia , Síndrome Metabólica/metabolismo , Camundongos Mutantes
7.
Clin Obstet Gynecol ; 56(3): 602-9, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23722920

RESUMO

Fetal glucocorticoid overexposure is a key potential mechanism underlying the link between low birthweight and later life diseases. The fetus is protected from high maternal glucocorticoid levels by the placental enzyme 11ß-hydroxysteroid dehydrogenase type 2. Antenatal glucocorticoid administration to women at threat of preterm labor, and high endogenous maternal glucocorticoid levels during pregnancy associate with lower birthweight. Long-term consequences for offspring include hypothalamic-pituitary-adrenal axis activation, increased metabolic and cardiovascular disorders, and neurodevelopmental sequelae. Strategies are needed to limit antenatal glucocorticoid use to those most at risk of preterm labor and to identify those most at risk of future disease.


Assuntos
11-beta-Hidroxiesteroide Desidrogenase Tipo 2/fisiologia , Doenças Cardiovasculares/embriologia , Dislipidemias/embriologia , Glucocorticoides/efeitos adversos , Hiperglicemia/embriologia , Sistema Hipotálamo-Hipofisário/efeitos dos fármacos , Sistema Hipófise-Suprarrenal/efeitos dos fármacos , Peso ao Nascer/efeitos dos fármacos , Doenças Cardiovasculares/induzido quimicamente , Dislipidemias/induzido quimicamente , Feminino , Desenvolvimento Fetal/efeitos dos fármacos , Humanos , Hiperglicemia/induzido quimicamente , Recém-Nascido , Trabalho de Parto Prematuro , Gravidez , Efeitos Tardios da Exposição Pré-Natal
8.
Exp Diabetes Res ; 2012: 565160, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23133443

RESUMO

The incidence of gestational diabetes is increasing worldwide, exposing large numbers of infants to hyperglycaemia whilst in utero. This exposure may have a long-term negative impact on the cardiovascular health of the offspring. Novel methods to assess cardiovascular status in the neonatal period are now available-including measuring arterial intima-media thickness and retinal photography. These measures will allow researchers to assess the relative impact of intrauterine exposures, distinguishing these from genetic or postnatal environmental factors. Understanding the long-term impact of the intrauterine environment should allow the development of more effective health policy and interventions to decrease the future burden of cardiovascular disease. Initiating disease prevention aimed at the developing fetus during the antenatal period may optimise community health outcomes.


Assuntos
Doenças Cardiovasculares/etiologia , Diabetes Mellitus Tipo 2/fisiopatologia , Diabetes Gestacional/fisiopatologia , Saúde da Família , Desenvolvimento Fetal , Hiperglicemia/embriologia , Gravidez em Diabéticas/fisiopatologia , Animais , Aterosclerose/epidemiologia , Aterosclerose/etiologia , Doenças Cardiovasculares/epidemiologia , Diabetes Mellitus Tipo 2/sangue , Diabetes Gestacional/sangue , Suscetibilidade a Doenças , Feminino , Humanos , Hiperglicemia/etiologia , Hiperglicemia/fisiopatologia , Masculino , Troca Materno-Fetal , Obesidade/epidemiologia , Obesidade/etiologia , Obesidade/fisiopatologia , Gravidez , Gravidez em Diabéticas/sangue , Risco
9.
Am J Pathol ; 180(1): 153-64, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22056361

RESUMO

Pregestational diabetes retards early embryonic growth. Placental and fetal growth are closely associated, suggesting that placental growth is also impaired. During the first trimester of gestation, oxygen tension rises steeply, leading to excessive production of reactive oxygen species (ROS), which is exacerbated in diabetes and may affect placental development. We hypothesized that oxygen modifies hyperglycemic effects on ROS formation, resulting in decreased first-trimester trophoblast growth. This was tested using a first trimester trophoblast-derived cell line (ACH-3P). Normoglycemia did not alter ACH-3P proliferation at 2.5%, 8%, and 21% oxygen. Hyperglycemic conditions for up to 3 days reduced cell number by 65% and resulted in cell cycle (G(1)- and S-phase) changes but only at 21% oxygen. Proliferation reduction could be partially restored by an inhibitor of mitogen-activated protein kinase (MAPK) ERK1/2 but not of Akt/PkB. Intracellular ROS elevation under hyperglycemia was oxygen independent, whereas mitochondrial superoxide levels were enhanced under hyperglycemia only at 21% oxygen. Intervention to modulate cytosolic and mitochondrial ROS, using ROS formation inducers and inhibitors, did not alter cell growth under hyperglycemia at 21% oxygen. The combination of hyperglycemia and high oxygen levels (21%) reduces proliferation of human first-trimester trophoblasts in a ROS-independent manner involving MAPK. This may account for reduced placental growth and, therefore, also for embryonic growth during the first-trimester pregestational diabetic pregnancies when the oxygen tension increases.


Assuntos
Diabetes Gestacional/fisiopatologia , Hiperglicemia/embriologia , Oxigênio/farmacologia , Espécies Reativas de Oxigênio/metabolismo , Trofoblastos/fisiologia , Antimetabólitos/farmacologia , Antioxidantes/farmacologia , Proliferação de Células , Células Cultivadas , Diabetes Gestacional/metabolismo , Inibidores Enzimáticos/farmacologia , Feminino , Humanos , Peróxido de Hidrogênio/metabolismo , Hiperglicemia/metabolismo , Hiperglicemia/fisiopatologia , Sistema de Sinalização das MAP Quinases/fisiologia , Mitocôndrias/enzimologia , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Gravidez , Primeiro Trimestre da Gravidez , Inibidores de Proteínas Quinases/farmacologia , Trofoblastos/metabolismo , Regulação para Cima
10.
Folia Histochem Cytobiol ; 49(2): 325-34, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21744335

RESUMO

In various tissues, glucocorticoids (GCs) are known to downregulate glucose transport systems; however, their effects on glucose transporters (GLUTs) in the placenta of a diabetic rat are unknown. Glucocorticoid hormone action within the cell is regulated by the glucocorticoid receptor (GR). Thus, this study was designed to investigate the relationship between GR and glucose transporter expression in the placenta of the diabetic rat. Our immunohistochemical results indicated that GR and glucose transporter protein 1 (GLUT 1) are expressed ubiquitously in the trophoblast and endothelial cells of the labyrinthine zone, where maternal fetal transport takes place in the rat placenta. Expression of GR in the junctional zone of the rat placenta was detected in giant cells, and in some spongiotrophoblast cells, but not in the glycogen cells. GLUT 1 was present, especially in glycogen cells during early pregnancy, and in the spongiotrophoblast cells of the junctional zone during late pregnancy. Amounts of GR and GLUT 1 protein were increased towards the end of gestation both in the control and the diabetic placenta. However, at days 17 and 19 of gestation, only the placental GR protein was significantly increased in the streptozotocin-induced diabetic rats compared to control rats. Diabetes led to a significant decrease in placental weight at gestation day 15. In contrast, at gestational days 17 and 21, the weights of the diabetic placenta were significantly increased as compared with the controls. Moreover, diabetes induced fetus intrauterine growth retardation at gestational days 13, 17 and 21. In conclusion, the localization pattern of GR and GLUT 1 proteins in the same cell types led us to believe that there might be a relationship between GR and GLUT 1 expressions at the cellular level. GLUT 1 does not play a pivotal role in diabetic pregnancies. However, placental growth abnormalities during diabetic pregnancy may be related to the amount of GR.


Assuntos
Diabetes Mellitus Experimental/embriologia , Diabetes Mellitus Experimental/metabolismo , Transportador de Glucose Tipo 1/metabolismo , Placenta/embriologia , Placenta/metabolismo , Receptores de Glucocorticoides/metabolismo , Animais , Glicemia/metabolismo , Peso Corporal , Diabetes Mellitus Experimental/sangue , Diabetes Mellitus Experimental/complicações , Feminino , Feto/metabolismo , Hiperglicemia/sangue , Hiperglicemia/complicações , Hiperglicemia/embriologia , Immunoblotting , Imuno-Histoquímica , Placenta/patologia , Gravidez , Transporte Proteico , Ratos , Estreptozocina
11.
Reproduction ; 141(5): 607-15, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21367963

RESUMO

Autophagy is critical to the process of development because mouse models have shown that lack of autophagy leads to developmental arrest during the pre-implantation stage of embryogenesis. The process of autophagy is regulated through signaling pathways, which respond to the cellular environment. Therefore, any alteration in the environment may lead to the dysregulation of the autophagic process potentially resulting in cell death. Using both in vitro and in vivo models to study autophagy in the pre-implantation murine embryo, we observed that the cells respond to environmental stressors (i.e. hyperglycemic environment) by increasing activation of autophagy in a differential pattern within the embryo. This upregulation is accompanied by an increase in apoptosis, which appears to plateau at high concentrations of glucose. The activation of the autophagic pathway was further confirmed by an increase in GAPDH activity in both in vivo and in vitro hyperglycemic models, which has been linked to autophagy through the activation of the Atg12 gene. Furthermore, this increase in autophagy in response to a hyperglycemic environment was observed as early as the oocyte stage. In conclusion, in this study, we provided evidence for a differential response of elevated activation of autophagy in embryos and oocytes exposed to a hyperglycemic environment.


Assuntos
Autofagia , Blastocisto/metabolismo , Glucose/metabolismo , Hiperglicemia/metabolismo , Transdução de Sinais , Estresse Fisiológico , Animais , Blastocisto/patologia , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Experimental/patologia , Técnicas de Cultura Embrionária , Desenvolvimento Embrionário , Feminino , Gliceraldeído-3-Fosfato Desidrogenases/metabolismo , Hiperglicemia/embriologia , Hiperglicemia/patologia , Camundongos , Oócitos/metabolismo , Oócitos/ultraestrutura , Regulação para Cima
13.
Birth Defects Res A Clin Mol Teratol ; 88(10): 791-803, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20890938

RESUMO

This article provides an overview of the rationale for diabetes preconception care interventions for women with diabetes and the efficacy in reducing the excess occurrence of major congenital malformations. The problems with broad use of individualized preconception care are considered. In addition, suggestions are made for the implementation of more comprehensive interventions in the community and usual diabetes care settings, to address the multiple ongoing challenges in the prevention of structural anomalies associated with preexisting diabetes. Based on the published evidence, successful preconception care can be considered to include: achievement of individualized target standardized glycosylated hemoglobin levels, adequate nutrition, and minimizing hypoglycemia before and after discontinuing effective contraception and during the transition to early prenatal care.


Assuntos
Anormalidades Congênitas/embriologia , Anormalidades Congênitas/prevenção & controle , Hiperglicemia/metabolismo , Cuidado Pré-Concepcional , Gravidez em Diabéticas , Diabetes Mellitus/metabolismo , Feminino , Hemoglobinas Glicadas/normas , Humanos , Hiperglicemia/embriologia , Hipoglicemia/metabolismo , Gravidez , Cuidado Pré-Natal , Teratogênicos/metabolismo , Mulheres
14.
Clin Sci (Lond) ; 114(5): 381-92, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-17927565

RESUMO

Substantial evidence suggests that poor intrauterine milieu elicited by maternal nutritional disturbance may programme susceptibility in the fetus to later development of chronic diseases, such as obesity, hypertension, cardiovascular disease and diabetes. One of the most interesting features of fetal programming is the evidence from several studies that the consequences may not be limited to the first-generation offspring and that it can be passed transgenerationally. In the present study, female rats (F0) were fed either a normal-protein diet [control diet (C); 19 g of protein/100 g of diet] or a low-protein diet [restricted diet (R); 5 g of protein/100 g of diet]. The offspring were termed according to the period and the types of diet the dams were fed, i.e. CC, RC, CR and RR (first letter indicates the diet during gestation and the second the diet during lactation). At 3 months of age, F1 females were bred to proven males, outside the experiment, to produce F2 offspring. At weaning, F2 offspring were divided by gender. RC1 offspring (with the number indicating the filial generation) were born with low birthweight, but afterwards they had catch-up growth, reaching the weight of the CC1 offspring. The increased glycaemia in RC1 offspring was associated with insulin resistance. CR1 and RR1 offspring had impaired growth with no changes in glucose metabolism. RC2 offspring had high BM (body mass) at birth, which was sustained over the whole experiment in male offspring. The F2 generation had more alteration in glucose metabolism than the F1 generation. CR2 and RC2 offspring had hyperglycaemia accompanied by hyperinsulinaemia and insulin resistance in both genders. CR2 offspring had an increase in body adiposity with hyperleptinaemia. In conclusion, low protein during gestation improves BM, fat mass and growth rate in F1 rats, but has adverse effects on glucose and leptin metabolism, resulting in insulin resistance in adult F1 and F2 offspring. Low protein during lactation has adverse effects on glucose, insulin and leptin metabolism, resulting in insulin resistance in adult F2 offspring. These findings suggest that low protein during gestation and/or lactation can be passed transgenerationally to the second generation.


Assuntos
Glicemia/metabolismo , Dieta com Restrição de Proteínas , Transtornos da Nutrição Fetal/fisiopatologia , Lactação/fisiologia , Efeitos Tardios da Exposição Pré-Natal/fisiopatologia , Animais , Biometria , Feminino , Desenvolvimento Fetal/fisiologia , Crescimento/fisiologia , Hiperglicemia/embriologia , Hiperglicemia/genética , Hiperinsulinismo/embriologia , Hiperinsulinismo/genética , Resistência à Insulina/genética , Resistência à Insulina/fisiologia , Leptina/sangue , Masculino , Gravidez , Fenômenos Fisiológicos da Nutrição Pré-Natal/fisiologia , Ratos
15.
Clin Sci (Lond) ; 113(5): 219-32, 2007 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-17663659

RESUMO

Compelling epidemiological evidence suggests that exposure to an adverse intrauterine environment, manifested by low-birth weight, is associated with cardiometabolic and behavioural disorders in adulthood. These observations have led to the concept of 'fetal programming'. The molecular mechanisms that underlie this relationship remain unclear, but are being extensively investigated using a number of experimental models. One major hypothesis for early life physiological programming implicates fetal overexposure to stress (glucocorticoid) hormones. Several animal studies have shown that prenatal glucocorticoid excess, either from endogenous overproduction with maternal stress or through exogenous administration to the mother or fetus, reduces birth weight and causes lifelong hypertension, hyperglycaemia and behavioural abnormality in the offspring. Intriguingly, these effects are transmitted across generations without further exposure to glucocorticoids, which suggests an epigenetic mechanism. These animal observations could have huge implications if extrapolated to humans, where glucocorticoids have extensive therapeutic use in obstetric and neonatal practice.


Assuntos
Desenvolvimento Fetal/fisiologia , Glucocorticoides/fisiologia , Efeitos Tardios da Exposição Pré-Natal , Estresse Psicológico , Adulto , Feminino , Humanos , Hiperglicemia/embriologia , Hipertensão/embriologia , Recém-Nascido de Baixo Peso , Recém-Nascido , Masculino , Gravidez
17.
Diabet Med ; 23(11): 1213-5, 2006 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17054597

RESUMO

AIM: Type 2 diabetes is frequently familial. Hyperglycaemia in pregnancy might act in addition to genetic factors to cause diabetes in the children of mothers with gestational diabetes mellitus (GDM). The first manifestation of this in female offspring is likely to be GDM in their own pregnancies. We compared the incidence of GDM in daughters of diabetic mothers and diabetic fathers to determine if in utero exposure to hyperglycaemia increased the risk of a diabetes-prone phenotype in offspring. METHODS: We analysed the outcome of a GDM screening programme in women with a family history of diabetes in their mother (n = 535), father (n = 566), both parents (n = 77) or neither (n = 4672). RESULTS: GDM was twice as common in the daughters of diabetic mothers (11%) than diabetic fathers (5%, P = 0.002). Women with two diabetic parents were no more likely to have GDM than women with only a diabetic mother. CONCLUSIONS: Genetic predisposition to GDM should be equally shared by daughters of diabetic mothers and fathers. An excess of maternal transmission of diabetes is consistent with an epigenetic effect of hyperglycaemia in pregnancy acting in addition to genetic factors to produce diabetes in the next generation.


Assuntos
Diabetes Mellitus Tipo 2/embriologia , Diabetes Gestacional/genética , Diabetes Mellitus Tipo 2/genética , Pai , Feminino , Humanos , Hiperglicemia/embriologia , Hiperglicemia/genética , Masculino , Mães , Linhagem , Gravidez , Efeitos Tardios da Exposição Pré-Natal , Fatores de Risco
19.
Diabetologia ; 49(6): 1412-20, 2006 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-16570165

RESUMO

AIMS/HYPOTHESIS: Prenatal glucocorticoid exposure causes lifelong hyperglycaemia in rat offspring, associated with permanently increased hepatic phosphoenolpyruvate carboxykinase 2 (PCK2), the rate-controlling enzyme of gluconeogenesis. To elucidate the mechanisms underlying the 'programming' of PCK2, this study examined the effect of prenatal dexamethasone treatment on expression of transcription factors that regulate Pck2. MATERIALS AND METHODS: Real-time RT-PCR and in situ hybridisation were used to measure and localise hepatic mRNA transcribed from the genes for PCK2, hepatocyte nuclear factor 4, alpha (HNF4A), transcription factor 1 (TCF1), CCAAT/enhancer binding protein, alpha (CEBPA), CEBPB, the glucocorticoid receptor (NR3C1) and peroxisome proliferative activated receptor, gamma, coactivator 1 alpha (PPARGC1A) in foetal and adult offspring of dams treated with dexamethasone or vehicle during the last week of gestation. RESULTS: Prenatal dexamethasone exposure significantly elevated Hnf4a mRNA expression in foetal and adult liver. This resulted from increased expression of isoforms derived from the 'adult' (P1) Hnf4a promoter. In contrast, isoforms from the 'foetal' (P2) promoter were markedly suppressed by dexamethasone. Like Pck2, the increase in hepatic Hnf4a mRNA occurred exclusively in the periportal zone. Foetal Tcf1 expression was also increased by dexamethasone treatment, but this did not persist into adulthood. Prenatal dexamethasone did not affect the amounts of foetal and/or adult Cebpa, Cebpb, Nr3c1 or Ppargc1a mRNA. CONCLUSIONS/INTERPRETATION: Prenatal dexamethasone exposure caused a permanent increase in hepatic Hnf4a mRNA. This increase, which was associated with a premature switch from foetal to adult promoter predominance, was congruent with changes in Pck2 expression. These data suggest that HNF4A might mediate Pck2 overexpression and subsequent hyperglycaemia.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento , Fator 4 Nuclear de Hepatócito/genética , Hiperglicemia/embriologia , Fígado/embriologia , Animais , Sequência de Bases , Primers do DNA , Dexametasona/farmacologia , Desenvolvimento Embrionário , Feminino , Desenvolvimento Fetal , Glucocorticoides/farmacologia , Fator 4 Nuclear de Hepatócito/efeitos dos fármacos , Gravidez , RNA Mensageiro/genética , Ratos , Transcrição Gênica
20.
J Physiol ; 572(Pt 1): 17-24, 2006 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-16455683

RESUMO

Glucose is the principal energy substrate for the the fetus and is essential for normal fetal metabolism and growth. Fetal glucose metabolism is directly dependent on the fetal plasma glucose concentration. Fetal glucose utilization is augmented by insulin produced by the developing fetal pancreas in increasing amounts as gestation proceeds, which enhances glucose utilization among the insulin-sensitive tissues (skeletal muscle, liver, heart, adipose tissue) that increase in mass and thus glucose need during late gestation. Glucose-stimulated insulin secretion increases over gestation. Both insulin secretion and insulin action are affected by prevailing glucose concentrations and the amount and activity of tissue glucose transporters. In cases of intrauterine growth restriction (IUGR), fetal weight-specific tissue glucose uptake rates and glucose transporters are maintained or increased, while synthesis of amino acids into protein and corresponding insulin-IGF signal transduction proteins are decreased. These observations demonstrate the mixed phenotype of the IUGR fetus that includes enhanced glucose utilization capacity, but diminished protein synthesis and growth. Thus, the fetus has considerable capacity to adapt to changes in glucose supply by relatively common and understandable mechanisms that regulate fetal metabolism and growth and could underlie certain later life metabolic disorders such as insulin resistance, obesity and diabetes mellitus.


Assuntos
Desenvolvimento Fetal/fisiologia , Feto/metabolismo , Glucose/metabolismo , Hiperglicemia/embriologia , Hiperglicemia/metabolismo , Insulina/metabolismo , Adaptação Fisiológica/fisiologia , Homeostase/fisiologia , Humanos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA