Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
1.
Am J Physiol Endocrinol Metab ; 304(1): E14-22, 2013 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-23092912

RESUMO

Low birth weight is associated with an increased risk for developing type 2 diabetes and metabolic diseases. The placental capacity to supply nutrients and oxygen to the fetus represents the main determiner of fetal growth. However, few studies have investigated the effects of maternal diet on the placenta. We explored placental adaptive proteomic processes implicated in response to maternal undernutrition. Rat term placentas from 70% food-restricted (FR30) mothers were used for a proteomic screen. Placental mitochondrial functions were evaluated using molecular and functional approaches, and ATP production was measured. FR30 drastically reduced placental and fetal weights. FR30 placentas displayed 14 proteins that were differentially expressed, including several mitochondrial proteins. FR30 induced a marked increase in placental mtDNA content and changes in mitochondrial functions, including modulation of the expression of genes implicated in biogenesis and bioenergetic pathways. FR30 mitochondria showed higher oxygen consumption but failed to maintain their ATP production. Maternal undernutrition induces placental mitochondrial abnormalities. Although an increase in biogenesis and bioenergetic efficiency was noted, placental ATP level was reduced. Our data suggest that placental mitochondrial defects may be implicated in fetoplacental pathologies.


Assuntos
Restrição Calórica/efeitos adversos , Metabolismo Energético/fisiologia , Retardo do Crescimento Fetal/etiologia , Fenômenos Fisiológicos da Nutrição Materna , Mitocôndrias/fisiologia , Placenta/metabolismo , Animais , Eficiência/fisiologia , Feminino , Retardo do Crescimento Fetal/metabolismo , Masculino , Troca Materno-Fetal/fisiologia , Mitocôndrias/metabolismo , Placenta/fisiologia , Placenta/ultraestrutura , Circulação Placentária/fisiologia , Gravidez , Ratos , Ratos Wistar
2.
Am J Physiol Regul Integr Comp Physiol ; 297(5): R1516-25, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19759337

RESUMO

Mitochondrial dysfunction may be a long-term consequence of a poor nutritional environment during early life. Our aim was to investigate whether a maternal low-protein (LP) diet may program mitochondrial dysfunction in islets of adult progeny before glucose intolerance ensues. To address this, pregnant Wistar rats were fed isocaloric diets containing either 20% protein (control) or 8% protein (LP diet) throughout gestation. From birth, offspring received the control diet. The mitochondrial function was analyzed in islets of 3-mo-old offspring. Related to their basal insulin release, cultured islets from both male and female LP offspring presented a lower response to glucose challenge and a blunted ATP production compared with control offspring. The expression of malate dehydrogenase as well as the subunit 6 of the ATP synthase encoded by mitochondrial genome (mtDNA) was lower in these islets, reducing the capacity of ATP production through the Krebs cycle and oxidative phosphorylation. However, mtDNA content was unchanged in LP islets compared with control. Several consequences of protein restriction during fetal life were more marked in male offspring. Only LP males showed an increased reactive oxygen species production associated with a higher expression of mitochondrial subunits of the electron transport chain NADH-ubiquinone oxireductase subunit 4L, an overexpression of peroxisome proliferator-activated receptor-gamma and uncoupling protein-2, and a strongly reduced beta-cell mass. In conclusion, mitochondrial function is clearly altered in islets from LP adult offspring in a sex-specific manner. That may provide a cellular explanation for the earlier development of glucose intolerance in male than in female offspring of dams fed an LP diet.


Assuntos
Dieta com Restrição de Proteínas , Ilhotas Pancreáticas/fisiologia , Mitocôndrias/fisiologia , Prenhez/fisiologia , Efeitos Tardios da Exposição Pré-Natal/fisiopatologia , Caracteres Sexuais , Trifosfato de Adenosina/metabolismo , Animais , Glicemia/metabolismo , Peso Corporal/fisiologia , DNA Mitocondrial/metabolismo , Ingestão de Alimentos/fisiologia , Feminino , Insulina/sangue , Lactação/fisiologia , Masculino , Modelos Animais , Gravidez , Ratos , Ratos Wistar , Espécies Reativas de Oxigênio/metabolismo
3.
Adv Exp Med Biol ; 646: 125-31, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19536671

RESUMO

A substantial body of evidence suggests that a poor intrauterine milieu elicited by maternal nutritional disturbance, including maternal diabetes or placental insufficiency, may programme susceptibility in the fetus to later development of glucose intolerance and diabetes. Numerous data in animals have allowed possible mechanisms for programming to be proposed. This review of work in several animal models attempts to identify the cellular and molecular mechanisms at the level of the beta-cell and in the insulin sensitive tissues that are involved in the process of events leading to the pathology later in life.


Assuntos
Diabetes Mellitus Tipo 2/metabolismo , Intolerância à Glucose/metabolismo , Fenômenos Fisiológicos da Nutrição do Lactente , Insulina/metabolismo , Fenômenos Fisiológicos da Nutrição Materna , Efeitos Tardios da Exposição Pré-Natal/metabolismo , Animais , Diabetes Mellitus Tipo 2/complicações , Modelos Animais de Doenças , Feminino , Humanos , Lactente , Recém-Nascido , Masculino , Gravidez , Fenômenos Fisiológicos da Nutrição Pré-Natal , Ratos
4.
Int J Biochem Cell Biol ; 38(5-6): 913-22, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-16337425

RESUMO

A substantial body of evidence now suggests that poor intrauterine milieu elicited by maternal nutritional disturbance or placental insufficiency may programme susceptibility in the foetus to later develop chronic degenerative diseases, such as obesity, hypertension, cardiovascular diseases and diabetes. Further data showing the developmental programming of the metabolic syndrome are now available thanks to animal studies in which the foetal environment has been manipulated. This review examines the developmental programming of glucose intolerance by disturbed intrauterine metabolic condition in rats. It focuses on the alteration of the endocrine pancreas at birth. Long-term consequences, deterioration of glucose tolerance and even transgenerational effects are reported. Maternal protein, caloric restriction and diabetes during gestation/lactation lead to altered beta-cell mass. This review also tempts to identify cellular and molecular mechanisms involved in this process.


Assuntos
Pâncreas/embriologia , Efeitos Tardios da Exposição Pré-Natal , Animais , Diabetes Gestacional/fisiopatologia , Feminino , Humanos , Células Secretoras de Insulina/patologia , Ilhotas Pancreáticas/embriologia , Mitocôndrias/fisiologia , Gravidez , Deficiência de Proteína/complicações , Desnutrição Proteico-Calórica/complicações , Útero/irrigação sanguínea
5.
Metabolism ; 55(5): 642-9, 2006 May.
Artigo em Inglês | MEDLINE | ID: mdl-16631441

RESUMO

Evidence to support an association between early nutrition and the development of obesity in the rat is equivocal. In this study we have investigated the postnatal growth, glucose tolerance, and adipocyte function of the offspring from pregnant rats fed with diets containing either 20% or 8% protein during gestation. By 25 weeks of age, the female offspring of dams fed with the diet containing 8% protein had a significantly lower adult body weight due in part to a decrease in body fat. The peak concentration of insulin after oral administration of a glucose dose was significantly lower in both the male and female offspring of the dams fed with the diet containing 8% protein. However, the ability of insulin to stimulate lipogenesis or suppress lipolysis in fat cells isolated from the offspring was not influenced by the prenatal diet. Hepatic phosphoenolpyruvate carboxykinase activity was reduced in female offspring of dams fed with the diet containing 8% protein. These results show that adult body composition is determined during the prenatal period as a result of programming of the insulin axis. This metabolic programming influences hepatic metabolism; however, there is no evidence for a programmed change in adipocyte function.


Assuntos
Composição Corporal/efeitos dos fármacos , Proteínas Alimentares/farmacologia , Desenvolvimento Fetal/efeitos dos fármacos , Insulina/metabolismo , Efeitos Tardios da Exposição Pré-Natal , Adipócitos/metabolismo , Animais , Animais Recém-Nascidos , Glicemia/metabolismo , Composição Corporal/fisiologia , Peso Corporal/fisiologia , Proteínas Alimentares/metabolismo , Feminino , Desenvolvimento Fetal/fisiologia , Glucoquinase/metabolismo , Teste de Tolerância a Glucose , Fígado/enzimologia , Fígado/metabolismo , Masculino , Fosfoenolpiruvato Carboxiquinase (ATP)/metabolismo , Gravidez , Ratos , Ratos Long-Evans
6.
J Endocrinol ; 185(3): 457-65, 2005 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-15930172

RESUMO

We have evaluated the influence of oligofructose (OFS), a fermentable dietary fibre, on glucose homeostasis, insulin production and intestinal glucagon-like peptide-1 (GLP-1) in streptozotocin-treated diabetic rats. Male Wistar rats received either i.v. streptozotocin (STZ; 40 mg/kg) or vehicle (CT); one week later, they were fed for 6 weeks with either the standard diet (STZ-CT), or with a diet containing 10% oligofructose (STZ-OFS); both diets were available ad libitum. In a second set of experiments (duration 4 weeks), a supplemental group of food-restricted rats (STZ-Res) receiving a similar intake as CT rats, was added. OFS improved glucose tolerance and reduced food intake as compared with STZ-CT rats in both the post-prandial state and after an oral glucose tolerance test. After 6 weeks, portal and pancreatic insulin concentrations were doubled in STZ-OFS rats. Food restriction improved these parameters when compared with STZ-CT rats, but to a lesser extent than in the STZ-OFS group. We have shown that OFS treatment increased portal and colonic GLP-1(7-36) amide levels and doubled colonic proglucagon and prohormone convertase 1 mRNA levels; both OFS and food restriction lowered ileal GLP-1(7-36) amide levels as compared with levels in STZ-CT rats. We propose that OFS, through its fermentation in the colon, promotes the expression and secretion of colonic peptides, namely GLP-1(7-36) amide, with beneficial consequences on glycaemia, insulin secretion and hyperphagia in diabetic rats.


Assuntos
Glicemia/análise , Colo/metabolismo , Diabetes Mellitus Experimental/tratamento farmacológico , Oligossacarídeos/uso terapêutico , Fragmentos de Peptídeos/sangue , Actinas/genética , Animais , Colo/química , Diabetes Mellitus Experimental/metabolismo , Ingestão de Alimentos/efeitos dos fármacos , Fermentação , Privação de Alimentos , Glucagon/sangue , Glucagon/genética , Peptídeo 1 Semelhante ao Glucagon , Peptídeos Semelhantes ao Glucagon , Teste de Tolerância a Glucose , Insulina/análise , Insulina/sangue , Ilhotas Pancreáticas/química , Masculino , Fragmentos de Peptídeos/análise , Proglucagon , Pró-Proteína Convertase 1/genética , Precursores de Proteínas/sangue , Precursores de Proteínas/genética , RNA Mensageiro/análise , Ratos , Ratos Wistar , Reação em Cadeia da Polimerase Via Transcriptase Reversa
7.
PLoS One ; 7(11): e47986, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23144842

RESUMO

Males and females responses to gestational overnutrition set the stage for subsequent sex-specific differences in adult onset non communicable diseases. Placenta, as a widely recognized programming agent, contibutes to the underlying processes. According to our previous findings, a high-fat diet during gestation triggers sex-specific epigenetic alterations within CpG and throughout the genome, together with the deregulation of clusters of imprinted genes. We further investigated the impact of diet and sex on placental histology, transcriptomic and epigenetic signatures in mice. Both basal gene expression and response to maternal high-fat diet were sexually dimorphic in whole placentas. Numerous genes showed sexually dimorphic expression, but only 11 genes regardless of the diet. In line with the key role of genes belonging to the sex chromosomes, 3 of these genes were Y-specific and 3 were X-specific. Amongst all the genes that were differentially expressed under a high-fat diet, only 16 genes were consistently affected in both males and females. The differences were not only quantitative but remarkably qualitative. The biological functions and networks of genes dysregulated differed markedly between the sexes. Seven genes of the epigenetic machinery were dysregulated, due to effects of diet, sex or both, including the Y- and X-linked histone demethylase paralogues Kdm5c and Kdm5d, which could mark differently male and female epigenomes. The DNA methyltransferase cofactor Dnmt3l gene expression was affected, reminiscent of our previous observation of changes in global DNA methylation. Overall, this striking sexual dimorphism of programming trajectories impose a considerable revision of the current dietary interventions protocols.


Assuntos
Dieta Hiperlipídica/efeitos adversos , Epigênese Genética , Expressão Gênica , Placenta/metabolismo , Efeitos Tardios da Exposição Pré-Natal/genética , Animais , DNA (Citosina-5-)-Metiltransferases/genética , DNA (Citosina-5-)-Metiltransferases/metabolismo , Feminino , Regulação Enzimológica da Expressão Gênica , Histona Desmetilases , Masculino , Camundongos , Análise de Sequência com Séries de Oligonucleotídeos , Oxirredutases N-Desmetilantes/genética , Oxirredutases N-Desmetilantes/metabolismo , Placenta/fisiopatologia , Gravidez , Efeitos Tardios da Exposição Pré-Natal/metabolismo , Fenômenos Fisiológicos da Nutrição Pré-Natal , Caracteres Sexuais , Transcriptoma
8.
World J Diabetes ; 2(9): 149-57, 2011 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-21954419

RESUMO

Under-nutrition as well as over-nutrition during pregnancy has been associated with the development of adult diseases such as diabetes and obesity. Both epigenetic modifications and programming of the mitochondrial function have been recently proposed to explain how altered intrauterine metabolic environment may produce such a phenotype. This review aims to report data reported in several animal models of fetal malnutrition due to maternal low protein or low calorie diet, high fat diet as well as reduction in placental blood flow. We focus our overview on the ß cell. We highlight that, notwithstanding early nutritional events, mitochondrial dysfunctions resulting from different alteration by diet or gender are programmed. This may explain the higher propensity to develop obesity and diabetes in later life.

9.
Am J Clin Nutr ; 94(6 Suppl): 1846S-1852S, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21543546

RESUMO

Studies on fetal undernutrition have generated the hypothesis that fetal programming corresponds to an attempt of the fetus to adapt to adverse conditions encountered in utero. These adaptations would be beneficial if these conditions prevail later in life, but they become detrimental in the case of normal or plentiful nutrition and favor the appearance of the metabolic syndrome. In this article, the discussion is limited to the developmental programming of obesity and cardiovascular disorders caused by an early mismatched nutrition, particularly intrauterine growth retardation followed by postnatal catch-up growth. Selected data in humans are reviewed before evoking some mechanisms revealed or suggested by experiments in rodents. A variety of physiologic mechanisms are implicated in obesity programming, 2 of which are detailed. In some, but not all observations, hyperphagia resulting namely from perturbed development of the hypothalamic circuitry devoted to appetite regulation may contribute to obesity. Another contribution may be the developmental changes in the population of fat cell precursors in adipose tissue. Even if the link between obesity and cardiovascular disease is well established, alteration of blood pressure regulation may appear independently of obesity. A loss of diurnal variation in heart rate and blood pressure in adulthood has resulted from maternal undernutrition followed by postnatal overnutrition. Further research should clarify the effect of mismatched early nutrition on the development of brain centers regulating energy intake, energy expenditure, and circadian rhythms.


Assuntos
Doenças Cardiovasculares/metabolismo , Desenvolvimento Fetal , Desnutrição/metabolismo , Fenômenos Fisiológicos da Nutrição Materna , Obesidade/metabolismo , Tecido Adiposo/metabolismo , Animais , Regulação do Apetite , Doenças Cardiovasculares/etiologia , Ingestão de Energia , Metabolismo Energético , Feminino , Humanos , Desnutrição/complicações , Modelos Animais , Estado Nutricional , Obesidade/etiologia , Gravidez , Efeitos Tardios da Exposição Pré-Natal , Roedores
10.
J Nutr Biochem ; 22(10): 985-94, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21190832

RESUMO

Accumulating evidence has shown that maternal malnutrition increases the risk of metabolic disease in the progeny. We previously reported that prenatal exposure to a low-protein diet (LP) leads to mitochondrial dysfunction in pancreatic islets from adult rodent offspring that could relate physiological and cellular alterations due to early diet. We aim to determine whether mitochondrial dysfunction could be a common consequence of prenatal nutritional unbalances. Pregnant Wistar rats received either a global food restriction (GFR), consisting in the reduction by 50% of the normal daily food intake, or a high-fat diet (HF) throughout gestation. GFR or HF diet during pregnancy leads to a lack of increase in insulin release and ATP content in response to glucose stimulation in islets from 3-month-old male and female offspring. These similar consequences originated from impairment in either glucose sensing or glucose metabolism, depending on the type of early malnutrition and on the sex of the progeny. Indeed, the glucose transport across ß-cell membrane seemed compromised in female HF offspring, since GLUT-2 gene was markedly underexpressed. Additionally, for each progeny, consequences downstream the entry of glucose were also apparent. Expression of genes involved in glycolysis, TCA cycle and oxidative phosphorylations was altered in GFR and HF rats in a sex- and diet-dependent manner. Moreover, prenatal malnutrition affected the regulators of mitochondrial biogenesis, namely, PPAR coactivator 1 alpha (PGC-1α), since its expression was higher in islets from GFR rats. In conclusion, programming of mitochondrial dysfunction is a consequence of maternal malnutrition, which may predispose to glucose intolerance in the adult offspring.


Assuntos
Ilhotas Pancreáticas/metabolismo , Fenômenos Fisiológicos da Nutrição Materna , Mitocôndrias/fisiologia , Trifosfato de Adenosina/metabolismo , Animais , Glicemia/metabolismo , Peso Corporal , DNA Mitocondrial/metabolismo , Dieta Hiperlipídica , Feminino , Insulina/metabolismo , Gravidez , Ratos , Ratos Wistar , Espécies Reativas de Oxigênio/metabolismo
11.
PLoS One ; 6(10): e25576, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21991320

RESUMO

AIM: Glucocorticoids (GCs) take part in the direct control of cell lineage during the late phase of pancreas development when endocrine and exocrine cell differentiation occurs. However, other tissues such as the vasculature exert a critical role before that phase. This study aims to investigate the consequences of overexposure to exogenous glucocorticoids during different time-windows of gestation for the development of the fetal endocrine pancreas. METHODS: Pregnant Wistar rats received dexamethasone acetate in their drinking water (1 µg/ml) during the last week or throughout gestation. Fetuses and their pancreases were analyzed at day 15 and 21 of gestation. Morphometrical analysis was performed on pancreatic sections after immunohistochemistry techniques and insulin secretion was evaluated on fetal islets collected in vitro. RESULTS: Dexamethasone given the last week or throughout gestation reduced the beta-cell mass in 21-day-old fetuses by respectively 18% or 62%. This was accompanied by a defect in insulin secretion. The alpha-cell mass was reduced similarly. Neither islet vascularization nor beta-cell proliferation was affected when dexamethasone was administered during the last week, which was however the case when given throughout gestation. When given from the beginning of gestation, dexamethasone reduced the number of cells expressing the early marker of endocrine lineage neurogenin-3 when analyzed at 15 days of fetal age. CONCLUSIONS: GCs reduce the beta- and alpha-cell mass by different mechanisms according to the stage of development during which the treatment was applied. In fetuses exposed to glucocorticoids the last week of gestation only, beta-cell mass is reduced due to impairment of beta-cell commitment, whereas in fetuses exposed throughout gestation, islet vascularization and lower beta-cell proliferation are involved as well, amplifying the reduction of the endocrine mass.


Assuntos
Dexametasona/farmacologia , Desenvolvimento Fetal/efeitos dos fármacos , Células Secretoras de Insulina/efeitos dos fármacos , Células Secretoras de Insulina/patologia , Exposição Materna , Animais , Apoptose/efeitos dos fármacos , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Glicemia/efeitos dos fármacos , Bromodesoxiuridina/metabolismo , Proliferação de Células/efeitos dos fármacos , Comportamento Alimentar/efeitos dos fármacos , Feminino , Feto/irrigação sanguínea , Feto/efeitos dos fármacos , Feto/patologia , Insulina/sangue , Insulina/metabolismo , Secreção de Insulina , Masculino , Neovascularização Fisiológica/efeitos dos fármacos , Proteínas do Tecido Nervoso/metabolismo , Tamanho do Órgão/efeitos dos fármacos , Ratos , Ratos Wistar , Fatores de Tempo , Fatores de Transcrição/metabolismo , Aumento de Peso/efeitos dos fármacos
12.
Am J Clin Nutr ; 94(6 Suppl): 1824S-1829S, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21562089

RESUMO

Type 2 diabetes arises when the endocrine pancreas fails to secrete sufficient insulin to cope with metabolic demands resulting from ß cell secretory dysfunction, decreased ß cell mass, or both. Epidemiologic studies have shown strong relations between poor fetal and early postnatal nutrition and susceptibility to diabetes later in life. Animal models have been established, and studies have shown that a reduction in the availability of nutrients during fetal development programs the endocrine pancreas and insulin-sensitive tissues. We investigated several modes of early malnutrition in rats. Regardless of the type of diet investigated, whether there was a deficit in calories or protein in food or even in the presence of a high-fat diet, malnourished pups were born with a defect in their ß cell population, with fewer ß cells that did not secrete enough insulin and that were more vulnerable to oxidative stress; such populations of ß cells will never completely recover. Despite the similar endpoint, the cellular and physiologic mechanisms that contribute to alterations in ß cell mass differ depending on the nature of the nutritional insult. Hormones that are operative during fetal life, such as insulin, insulin-like growth factors, and glucocorticoids; specific molecules, such as taurine; and islet vascularization have been implicated as possible factors in amplifying this defect. The molecular mechanisms responsible for intrauterine programming of ß cells are still elusive, but among them the programming of mitochondria may be a strong central candidate.


Assuntos
Desenvolvimento Fetal , Ilhotas Pancreáticas/embriologia , Desnutrição/patologia , Fenômenos Fisiológicos da Nutrição Materna , Efeitos Tardios da Exposição Pré-Natal/patologia , Animais , DNA Mitocondrial/genética , DNA Mitocondrial/metabolismo , Diabetes Mellitus Tipo 2/embriologia , Diabetes Mellitus Tipo 2/etiologia , Diabetes Mellitus Tipo 2/fisiopatologia , Dieta Hiperlipídica , Dieta com Restrição de Proteínas , Modelos Animais de Doenças , Determinação de Ponto Final , Epigênese Genética , Feminino , Regulação da Expressão Gênica , Predisposição Genética para Doença , Humanos , Recém-Nascido , Doenças do Recém-Nascido/etiologia , Doenças do Recém-Nascido/patologia , Insulina/metabolismo , Secreção de Insulina , Células Secretoras de Insulina/citologia , Células Secretoras de Insulina/metabolismo , Ilhotas Pancreáticas/efeitos dos fármacos , Desnutrição/complicações , Troca Materno-Fetal , Gravidez , Efeitos Tardios da Exposição Pré-Natal/etiologia , Ratos , Somatomedinas/metabolismo
13.
PLoS One ; 5(9)2010 Sep 09.
Artigo em Inglês | MEDLINE | ID: mdl-20844591

RESUMO

BACKGROUND: A link between early mismatched nutritional environment and development of components of the metabolic syndrome later in life has been shown in epidemiological and animal data. The aim of this study was to investigate whether an early mismatched nutrition produced by catch-up growth after fetal protein restriction could induce the appearance of hypertension and/or atherosclerosis in adult male mice. METHODOLOGY/PRINCIPAL FINDINGS: Wild-type C57BL6/J or LDLr-/- dams were fed a low protein (LP) or a control (C) diet during gestation. Catch-up growth was induced in LP offspring by feeding dams with a control diet and by culling the litter to 4 pups against 8 in controls. At weaning, male mice were fed either standard chow or an obesogenic diet (OB), leading to 4 experimental groups. Blood pressure (BP) and heart rate (HR) were assessed in conscious unrestrained wild-type mice by telemetry. Atherosclerosis plaque area was measured in aortic root sections of LDLr-/- mice. We found that: (1) postnatal OB diet increased significantly BP (P<0.0001) and HR (P<0.008) in 3-month old OB-C and OB-LP offspring, respectively; (2) that maternal LP diet induced a significant higher BP (P<0.009) and HR (P<0.004) and (3) an altered circadian rhythm in addition to higher plasma corticosterone concentration in 9 months-old LP offspring; (4) that, although LP offspring showed higher plasma total cholesterol than control offspring, atherosclerosis assessed in aortic roots of 6-mo old mice featured increased plaque area due to OB feeding but not due to early mismatched nutrition. CONCLUSIONS/SIGNIFICANCE: These results indicate a long-term effect of early mismatched nutrition on the appearance of hypertension independently of obesity, while no effect on atherosclerosis was noticed at this age.


Assuntos
Aterosclerose/etiologia , Dieta com Restrição de Proteínas/efeitos adversos , Suscetibilidade a Doenças , Hipertensão/etiologia , Fenômenos Fisiológicos da Nutrição Materna , Efeitos Tardios da Exposição Pré-Natal , Animais , Aterosclerose/fisiopatologia , Pressão Sanguínea , Modelos Animais de Doenças , Feminino , Frequência Cardíaca , Humanos , Hipertensão/fisiopatologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Gravidez , Especificidade da Espécie
14.
PLoS One ; 5(12): e14398, 2010 Dec 21.
Artigo em Inglês | MEDLINE | ID: mdl-21200436

RESUMO

BACKGROUND: Changes in imprinted gene dosage in the placenta may compromise the prenatal control of nutritional resources. Indeed monoallelic behaviour and sensitivity to changes in regional epigenetic state render imprinted genes both vulnerable and adaptable. METHODS AND FINDINGS: We investigated whether a high-fat diet (HFD) during pregnancy modified the expression of imprinted genes and local and global DNA methylation patterns in the placenta. Pregnant mice were fed a HFD or a control diet (CD) during the first 15 days of gestation. We compared gene expression patterns in total placenta homogenates, for male and female offspring, by the RT-qPCR analysis of 20 imprinted genes. Sexual dimorphism and sensitivity to diet were observed for nine genes from four clusters on chromosomes 6, 7, 12 and 17. As assessed by in situ hybridization, these changes were not due to variation in the proportions of the placental layers. Bisulphite-sequencing analysis of 30 CpGs within the differentially methylated region (DMR) of the chromosome 17 cluster revealed sex- and diet-specific differential methylation of individual CpGs in two conspicuous subregions. Bioinformatic analysis suggested that these differentially methylated CpGs might lie within recognition elements or binding sites for transcription factors or factors involved in chromatin remodelling. Placental global DNA methylation, as assessed by the LUMA technique, was also sexually dimorphic on the CD, with lower methylation levels in male than in female placentae. The HFD led to global DNA hypomethylation only in female placenta. Bisulphite pyrosequencing showed that neither B1 nor LINE repetitive elements could account for these differences in DNA methylation. CONCLUSIONS: A HFD during gestation triggers sex-specific epigenetic alterations within CpG and throughout the genome, together with the deregulation of clusters of imprinted genes important in the control of many cellular, metabolic and physiological functions potentially involved in adaptation and/or evolution. These findings highlight the importance of studying both sexes in epidemiological protocols and dietary interventions.


Assuntos
Ração Animal , Metilação de DNA , Gorduras na Dieta/metabolismo , Regulação da Expressão Gênica , Impressão Genômica , Placenta/metabolismo , Alelos , Animais , Epigênese Genética , Feminino , Dosagem de Genes , Masculino , Camundongos , Gravidez , Prenhez , Fatores Sexuais
15.
PLoS One ; 4(7): e6110, 2009 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-19568427

RESUMO

BACKGROUND: Islets from adult rat possess weak antioxidant defense leading to unbalance between superoxide dismutase (SOD) and hydrogen peroxide-inactivating enzymatic activities, catalase (CAT) and glutathione peroxidase (GPX) rending them susceptible to oxidative stress. We have shown that this vulnerability is influenced by maternal diet during gestation and lactation. METHODOLOGY/PRINCIPAL FINDINGS: The present study investigated if low antioxidant activity in islets is already observed at birth and if maternal protein restriction influences the development of islet antioxidant defenses. Rats were fed a control diet (C group) or a low protein diet during gestation (LP) or until weaning (LPT), after which offspring received the control diet. We found that antioxidant enzymatic activities varied with age. At birth and after weaning, normal islets possessed an efficient GPX activity. However, the antioxidant capacity decreased thereafter increasing the potential vulnerability to oxidative stress. Maternal protein malnutrition changed the antioxidant enzymatic activities in islets of the progeny. At 3 months, SOD activity was increased in LP and LPT islets with no concomitant activation of CAT and GPX. This unbalance could lead to higher hydrogen peroxide production, which may concur to oxidative stress causing defective insulin gene expression due to modification of critical factors that modulate the insulin promoter. We found indeed that insulin mRNA level was reduced in both groups of malnourished offspring compared to controls. Analyzing the expression of such critical factors, we found that c-Myc expression was strongly increased in islets from both protein-restricted groups compared to controls. CONCLUSION AND SIGNIFICANCE: Modification in antioxidant activity by maternal low protein diet could predispose to pancreatic islet dysfunction later in life and provide new insights to define a molecular mechanism responsible for intrauterine programming of endocrine pancreas.


Assuntos
Antioxidantes/metabolismo , Proteínas Alimentares/administração & dosagem , Ilhotas Pancreáticas/metabolismo , Animais , Peso Corporal , Catalase/metabolismo , Ativação Enzimática , Genes myc , Glutationa Peroxidase/metabolismo , Insulina/genética , Ilhotas Pancreáticas/enzimologia , Tamanho da Ninhada de Vivíparos , Peroxirredoxinas/genética , Ratos
16.
Obesity (Silver Spring) ; 16(12): 2760-3, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-18833213

RESUMO

We studied the in vitro proliferation and differentiation of rat preadipocytes to investigate whether catch-up growth after prenatal protein restriction may program adipose precursor cells leading to development of increased adipose tissue mass. Pregnant rat dams were fed either an isocaloric low-protein diet (LP-8%) or control diet (C-20%). During lactation, in order to induce catch-up growth, dams from LP group were fed with the C diet and litter size was reduced to four pups instead of eight. Preadipocytes were isolated from weanling male pups (28 days of age). Differentiation and proliferation were assessed across time. At late stages of preadipocyte differentiation, no difference was observed in lipid accumulation of C or LP cultures but the mRNA expression of leptin was enhanced in LP cells. At early stages of culture, a higher DNA and protein content accompanied by a higher rate of proliferation was measured in adipocytes from LP cultures. Moreover, the mRNA expression of cyclin D1 was increased in these cells whereas the expression of peroxisome proliferators-activated receptor gamma (PPARgamma) and steroyl regulatory element binding protein (SREBP-1c) was significantly reduced during early stages. The results suggest that prenatal exposure to a LP followed by rapid catch-up growth is associated with a higher rate for proliferation in preadipocytes.


Assuntos
Adipócitos/citologia , Tecido Adiposo/crescimento & desenvolvimento , Diferenciação Celular , Proliferação de Células , Dieta com Restrição de Proteínas , Crescimento , Adipócitos/metabolismo , Animais , Animais Recém-Nascidos , Ciclina D1/metabolismo , Feminino , Leptina/metabolismo , Masculino , PPAR gama/metabolismo , Gravidez , Proteínas/análise , RNA Mensageiro/metabolismo , Ratos , Proteína de Ligação a Elemento Regulador de Esterol 1/metabolismo
17.
Curr Drug Targets ; 8(8): 935-41, 2007 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-17691930

RESUMO

Type 2 diabetes, which has dramatically increased during the last decade normally results from a combination of pancreatic beta cell dysfunction and insulin resistance. One of the most recent risk factors identified for type 2 diabetes is a sub-optimal fetal and neonatal environment. Numerous human epidemiological studies worldwide have highlighted that a disturbed nutritional environment of the fetus, either poor or too abundant will compromise the health of the offspring by increasing the susceptibility to insulin resistance, to glucose intolerance and to diabetes in later life. In addition to adverse intrauterine events, the detrimental role of catch-up growth and of the mismatch between the prenatal and the postnatal metabolic environment in such pathology is now clear. To understand the mechanisms that are responsible for such programming and to be able to design prevention strategies, a number of animal models have been created. This manuscript reviews the data from several rodent models in which maternal or neonatal diet has been altered. These include models of maternal under-nutrition and over-nutrition as well as gestational diabetes. In general, abnormal beta cell mass and beta cell dysfunction are present at birth and insulin resistance, glucose intolerance and diabetes appear in adult offspring. Obesity, pregnancy and ageing exaggerate the phenotype and there is some evidence to suggest that the phenotype can be transmitted to a second generation independently of any further environmental modification. Possible underlying mechanisms are discussed and evidence for potential early intervention strategies are reported.


Assuntos
Diabetes Mellitus Tipo 2/etiologia , Efeitos Tardios da Exposição Pré-Natal , Fenômenos Fisiológicos da Nutrição Pré-Natal , Adulto , Animais , Diabetes Mellitus Tipo 2/embriologia , Diabetes Mellitus Tipo 2/epidemiologia , Diabetes Mellitus Tipo 2/fisiopatologia , Diabetes Gestacional/fisiopatologia , Modelos Animais de Doenças , Feminino , Desenvolvimento Fetal , Humanos , Recém-Nascido , Distúrbios Nutricionais , Obesidade/complicações , Obesidade/fisiopatologia , Fenótipo , Gravidez , Fatores de Risco
18.
Curr Opin Clin Nutr Metab Care ; 9(6): 728-33, 2006 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17053427

RESUMO

PURPOSE OF REVIEW: Taurine, a free amino acid, is found in millimolar concentrations in most mammalian tissues. Mammals are able to synthesize taurine endogenously, but some species such as humans are more dependent on dietary sources of taurine. A growing body of evidence suggests that taurine plays a preponderant role in many physiological processes, which will be summarized in this review. RECENT FINDINGS: Evidence for the requirement of taurine in the human diet has been obtained in many studies involving animal models and a few clinical trials. Recent and past studies suggested that taurine might be a pertinent candidate for use as a nutritional supplement to protect against oxidative stress, neurodegenerative diseases or atherosclerosis. Taurine has demonstrated promising actions in vitro, and as a result clinical trials have begun to investigate its effects on various diseases. SUMMARY: Taurine appears to have multiple functions and plays an important role in many physiological processes, such as osmoregulation, immunomodulation and bile salt formation. Taurine analogues/derivatives have recently been reported to have a marked activity on various disorders. Taken together, these observations actualize the old story of taurine.


Assuntos
Necessidades Nutricionais , Taurina/administração & dosagem , Taurina/biossíntese , Animais , Diabetes Mellitus Tipo 2/prevenção & controle , Humanos , Fatores Imunológicos , Hepatopatias/prevenção & controle , Doenças do Sistema Nervoso/prevenção & controle , Especificidade da Espécie , Taurina/deficiência , Taurina/fisiologia , Equilíbrio Hidroeletrolítico/fisiologia
19.
Am J Physiol Endocrinol Metab ; 291(4): E835-42, 2006 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-16720626

RESUMO

During perinatal development, the regulation of IGF system appears to be growth hormone (GH) independent. By using highly purified primary fetal hepatocytes, we investigated the role of prolactin (PRL) in the regulation of IGF system and hepatocyte proliferation. We also analyzed the consequence of a maternal low-protein (LP) diet on the regulation of IGF, IGF-binding protein (IGFBP), and hepatocyte proliferation by prolactin. Pregnant Wistar rats were fed a control (C) diet (20% protein) or isocaloric (LP; 8%) diet throughout gestation. On day 21.5, fetal hepatocytes were cultured for 4 days and incubated with rat prolactin. In the C hepatocytes, PRL at 100 ng/ml decreased the abundance of IGFBP-1 and IGFBP-2 by 50 (P < 0.05) and 60% (P < 0.01), respectively. It also reduced by 70% the level of IGF-II mRNA (P < 0.01). By contrast, PRL failed to modulate IGFBP-1 and IGFBP-2 production by LP hepatocytes, and this was associated with reduced abundance of the short form of PRL receptor (P < 0.05). PRL had no effect on either the proliferation or the IGF-I production by C and LP hepatocytes, although it reduced the expression of IGF-II. These results suggest that prolactin influences hepatocyte proliferation in vitro by inhibiting IGFBP-1, IGFBP-2, and IGF-II levels, which may coincide with the decline of IGF-II observed in rodents during late gestation in vivo. On the other hand, maternal LP diet induces a resistance of fetal hepatocytes to PRL.


Assuntos
Proteína 1 de Ligação a Fator de Crescimento Semelhante à Insulina/metabolismo , Proteína 2 de Ligação a Fator de Crescimento Semelhante à Insulina/metabolismo , Fator de Crescimento Insulin-Like II/metabolismo , Fator de Crescimento Insulin-Like I/metabolismo , Fígado/metabolismo , Desnutrição/metabolismo , Prolactina/farmacologia , Animais , Western Blotting , Processos de Crescimento Celular/fisiologia , Meios de Cultivo Condicionados/metabolismo , Proteínas Alimentares/metabolismo , Feminino , Desenvolvimento Fetal/fisiologia , Hepatócitos/citologia , Hepatócitos/metabolismo , Proteína 1 de Ligação a Fator de Crescimento Semelhante à Insulina/antagonistas & inibidores , Proteína 2 de Ligação a Fator de Crescimento Semelhante à Insulina/antagonistas & inibidores , Fator de Crescimento Insulin-Like I/antagonistas & inibidores , Fator de Crescimento Insulin-Like II/antagonistas & inibidores , Fator de Crescimento Insulin-Like II/genética , Fígado/citologia , Fígado/embriologia , Masculino , Gravidez , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Distribuição Aleatória , Ratos , Ratos Wistar , Reação em Cadeia da Polimerase Via Transcriptase Reversa
20.
Obesity (Silver Spring) ; 14(8): 1330-43, 2006 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16988075

RESUMO

OBJECTIVE: To investigate whether catch-up growth after maternal malnutrition would favor the development of obesity in adulthood. RESEARCH METHODS AND PROCEDURES: Pregnant rats were submitted to protein or calorie restriction during the course of gestation. During lactation, pups were protein-restricted, normally fed, or overfed [reduced litter size, control (C) diet]. At weaning, rats were transferred to chow or to a hypercaloric diet (HCD) known to induce obesity. Body weight, food intake, blood parameters, glucose tolerance, adipocyte cellularity, and adipose factors contributing to cardiovascular disease development were measured. RESULTS: Protein and calorie restriction during gestation led to growth retardation at birth. If malnutrition was prolonged throughout lactation, adult body weight was permanently reduced. However, growth-retarded offspring overfed during the suckling period underwent a rapid catch-up growth and became heavier than the normally fed Cs. Offspring of calorie-restricted rats gained more weight than those of dams fed protein-restricted diet. Feeding an HCD postnatally amplified the effect of calorie restriction, and offspring that underwent catch-up growth became more obese than Cs. The HCD was associated with hyperphagia, hyperglycemia, hyperinsulinemia, glucose intolerance, insulin resistance, and adipocyte hypertrophy. The magnitude of effects varied depending on the type and the timing of early malnutrition. The expression of genes encoding factors implicated in cardiovascular disease was also modulated differently by early malnutrition and adult obesity. DISCUSSION: Catch-up growth immediately after early malnutrition should be a key point for the programming of obesity.


Assuntos
Peso Corporal/fisiologia , Obesidade/fisiopatologia , Efeitos Tardios da Exposição Pré-Natal , Desnutrição Proteico-Calórica/fisiopatologia , Adipócitos/citologia , Adipócitos/metabolismo , Adiponectina/genética , Tecido Adiposo/citologia , Tecido Adiposo/metabolismo , Angiotensinogênio/genética , Animais , Animais Recém-Nascidos , Dieta , Ingestão de Alimentos/fisiologia , Feminino , Leptina/sangue , Tamanho da Ninhada de Vivíparos , Masculino , Obesidade/sangue , Obesidade/etiologia , Inibidor 1 de Ativador de Plasminogênio/genética , Gravidez , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Ratos , Ratos Wistar , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fatores de Tempo , Triglicerídeos/sangue
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA