Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Am J Clin Nutr ; 103(2): 579-88, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26718412

RESUMO

BACKGROUND: It is well established that low birth weight and accelerated postnatal growth increase the risk of liver dysfunction in later life. However, molecular mechanisms underlying such developmental programming are not well characterized, and potential intervention strategies are poorly defined. OBJECTIVES: We tested the hypotheses that poor maternal nutrition and accelerated postnatal growth would lead to increased hepatic fibrosis (a pathological marker of liver dysfunction) and that postnatal supplementation with the antioxidant coenzyme Q10 (CoQ10) would prevent this programmed phenotype. DESIGN: A rat model of maternal protein restriction was used to generate low-birth-weight offspring that underwent accelerated postnatal growth (termed "recuperated"). These were compared with control rats. Offspring were weaned onto standard feed pellets with or without dietary CoQ10 (1 mg/kg body weight per day) supplementation. At 12 mo, hepatic fibrosis, indexes of inflammation, oxidative stress, and insulin signaling were measured by histology, Western blot, ELISA, and reverse transcriptase-polymerase chain reaction. RESULTS: Hepatic collagen deposition (diameter of deposit) was greater in recuperated offspring (mean ± SEM: 12 ± 2 µm) than in controls (5 ± 0.5 µm) (P < 0.001). This was associated with greater inflammation (interleukin 6: 38% ± 24% increase; P < 0.05; tumor necrosis factor α: 64% ± 24% increase; P < 0.05), lipid peroxidation (4-hydroxynonenal, measured by ELISA: 0.30 ± 0.02 compared with 0.19 ± 0.05 µg/mL per µg protein; P < 0.05), and hyperinsulinemia (P < 0.05). CoQ10 supplementation increased (P < 0.01) hepatic CoQ10 concentrations and ameliorated liver fibrosis (P < 0.001), inflammation (P < 0.001), some measures of oxidative stress (P < 0.001), and hyperinsulinemia (P < 0.01). CONCLUSIONS: Suboptimal in utero nutrition combined with accelerated postnatal catch-up growth caused more hepatic fibrosis in adulthood, which was associated with higher indexes of oxidative stress and inflammation and hyperinsulinemia. CoQ10 supplementation prevented liver fibrosis accompanied by downregulation of oxidative stress, inflammation, and hyperinsulinemia.


Assuntos
Anti-Inflamatórios não Esteroides/uso terapêutico , Suplementos Nutricionais , Retardo do Crescimento Fetal/dietoterapia , Hepatite/prevenção & controle , Cirrose Hepática/prevenção & controle , Estresse Oxidativo , Ubiquinona/análogos & derivados , Animais , Citocinas/antagonistas & inibidores , Citocinas/sangue , Citocinas/metabolismo , Dieta com Restrição de Proteínas/efeitos adversos , Feminino , Desenvolvimento Fetal , Retardo do Crescimento Fetal/etiologia , Retardo do Crescimento Fetal/imunologia , Retardo do Crescimento Fetal/fisiopatologia , Hepatite/etiologia , Hepatite/metabolismo , Hepatite/patologia , Hiperinsulinismo/etiologia , Hiperinsulinismo/prevenção & controle , Fígado/imunologia , Fígado/metabolismo , Fígado/patologia , Cirrose Hepática/etiologia , Cirrose Hepática/metabolismo , Cirrose Hepática/patologia , Masculino , Desnutrição/fisiopatologia , Fenômenos Fisiológicos da Nutrição Materna , Gravidez , Complicações na Gravidez/fisiopatologia , Ratos Wistar , Organismos Livres de Patógenos Específicos , Ubiquinona/uso terapêutico , Desmame
2.
Endocrinology ; 156(10): 3528-37, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26214037

RESUMO

Low birth weight and rapid postnatal growth increases the risk of developing insulin resistance and type 2 diabetes in later life. However, underlying mechanisms and potential intervention strategies are poorly defined. Here we demonstrate that male Wistar rats exposed to a low-protein diet in utero that had a low birth weight but then underwent postnatal catch-up growth (recuperated offspring) had reductions in the insulin signaling proteins p110-ß (13% ± 6% of controls [P < .001]) and insulin receptor substrate-1 (39% ± 10% of controls [P < .05]) in adipose tissue. These changes were not accompanied by any change in expression of the corresponding mRNAs, suggesting posttranscriptional regulation. Recuperated animals displayed evidence of a proinflammatory phenotype of their adipose tissue with increased IL-6 (139% ± 8% [P < .05]) and IL1-ß (154% ± 16% [P < .05]) that may contribute to the insulin signaling protein dysregulation. Postweaning dietary supplementation of recuperated animals with coenzyme Q (CoQ10) (1 mg/kg of body weight per day) prevented the programmed reduction in insulin receptor substrate-1 and p110-ß and the programmed increased in IL-6. These findings suggest that postweaning CoQ10 supplementation has antiinflammatory properties and can prevent programmed changes in insulin-signaling protein expression. We conclude that CoQ10 supplementation represents an attractive intervention strategy to prevent the development of insulin resistance that results from suboptimal in utero nutrition.


Assuntos
Inflamação/metabolismo , Resistência à Insulina , Insulina/metabolismo , Fenômenos Fisiológicos da Nutrição Materna , Transdução de Sinais , Ubiquinona/análogos & derivados , Tecido Adiposo/metabolismo , Animais , Feminino , Perfilação da Expressão Gênica , Transtornos do Crescimento/fisiopatologia , Insulina/sangue , Lipídeos/sangue , Masculino , Exposição Materna , Camundongos , MicroRNAs/metabolismo , Estresse Oxidativo , Fenótipo , Ratos , Ratos Wistar , Ubiquinona/fisiologia
3.
FASEB J ; 28(12): 5398-405, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25172893

RESUMO

Low birth weight and rapid postnatal growth increases risk of cardiovascular-disease (CVD); however, underlying mechanisms are poorly understood. Previously, we demonstrated that rats exposed to a low-protein diet in utero that underwent postnatal catch-up growth (recuperated) have a programmed deficit in cardiac coenzyme Q (CoQ) that was associated with accelerated cardiac aging. It is unknown whether this deficit occurs in all tissues, including those that are clinically accessible. We investigated whether aortic and white blood cell (WBC) CoQ is programmed by suboptimal early nutrition and whether postweaning dietary supplementation with CoQ could prevent programmed accelerated aging. Recuperated male rats had reduced aortic CoQ [22 d (35±8.4%; P<0.05); 12 m (53±8.8%; P<0.05)], accelerated aortic telomere shortening (P<0.01), increased DNA damage (79±13% increase in nei-endonucleaseVIII-like-1), increased oxidative stress (458±67% increase in NAPDH-oxidase-4; P<0.001), and decreased mitochondrial complex II-III activity (P<0.05). Postweaning dietary supplementation with CoQ prevented these detrimental programming effects. Recuperated WBCs also had reduced CoQ (74±5.8%; P<0.05). Notably, WBC CoQ levels correlated with aortic telomere-length (P<0.0001) suggesting its potential as a diagnostic marker of vascular aging. We conclude that early intervention with CoQ in at-risk individuals may be a cost-effective and safe way of reducing the global burden of CVDs.


Assuntos
Doenças Cardiovasculares/prevenção & controle , Suplementos Nutricionais , Ubiquinona/metabolismo , Animais , Doenças Cardiovasculares/enzimologia , Feminino , Estresse Oxidativo , Gravidez , Ratos Wistar , Telomerase/metabolismo , Ubiquinona/administração & dosagem
4.
Mol Metab ; 2(4): 480-90, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24327963

RESUMO

Studies in human and animals have demonstrated that nutritionally induced low birth-weight followed by rapid postnatal growth increases the risk of metabolic syndrome and cardiovascular disease. Although the mechanisms underlying such nutritional programming are not clearly defined, increased oxidative-stress leading to accelerated cellular aging has been proposed to play an important role. Using an established rodent model of low birth-weight and catch-up growth, we show here that post-weaning dietary supplementation with coenzyme Q10, a key component of the electron transport chain and a potent antioxidant rescued many of the detrimental effects of nutritional programming on cardiac aging. This included a reduction in nitrosative and oxidative-stress, telomere shortening, DNA damage, cellular senescence and apoptosis. These findings demonstrate the potential for postnatal antioxidant intervention to reverse deleterious phenotypes of developmental programming and therefore provide insight into a potential translatable therapy to prevent cardiovascular disease in at risk humans.

5.
PLoS One ; 5(4): e10074, 2010 Apr 09.
Artigo em Inglês | MEDLINE | ID: mdl-20404917

RESUMO

The negative impact of obesity on reproductive success is well documented but the stages at which development of the conceptus is compromised and the mechanisms responsible for the developmental failure still remain unclear. Recent findings suggest that mitochondria may be a contributing factor. However to date no studies have directly addressed the consequences of maternal obesity on mitochondria in early embryogenesis.Using an established murine model of maternal diet induced obesity and a live cell dynamic fluorescence imaging techniques coupled with molecular biology we have investigated the underlying mechanisms of obesity-induced reduced fertility. Our study is the first to show that maternal obesity prior to conception is associated with altered mitochondria in mouse oocytes and zygotes. Specifically, maternal diet-induced obesity in mice led to an increase in mitochondrial potential, mitochondrial DNA content and biogenesis. Generation of reactive oxygen species (ROS) was raised while glutathione was depleted and the redox state became more oxidised, suggestive of oxidative stress. These altered mitochondrial properties were associated with significant developmental impairment as shown by the increased number of obese mothers who failed to support blastocyst formation compared to lean dams. We propose that compromised oocyte and early embryo mitochondrial metabolism, resulting from excessive nutrient exposure prior to and during conception, may underlie poor reproductive outcomes frequently reported in obese women.


Assuntos
Dieta/efeitos adversos , Fenômenos Fisiológicos da Nutrição Materna , Mitocôndrias/metabolismo , Obesidade/metabolismo , Oócitos/metabolismo , Zigoto/metabolismo , Animais , Desenvolvimento Embrionário , Feminino , Fertilidade , Humanos , Camundongos , Modelos Animais , Mães , Obesidade/etiologia , Oxirredução , Estresse Oxidativo , Gravidez , Reprodução
6.
Hepatology ; 50(6): 1796-808, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19816994

RESUMO

UNLABELLED: Nonalcoholic fatty liver disease (NAFLD) describes an increasingly prevalent spectrum of liver disorders associated with obesity and metabolic syndrome. It is uncertain why steatosis occurs in some individuals, whereas nonalcoholic steatohepatitis (NASH) occurs in others. We have generated a novel mouse model to test our hypothesis: that maternal fat intake contributes to the development of NAFLD in adult offspring. Female mice were fed either a high-fat (HF) or control chow (C) diet before and during gestation and lactation. Resulting offspring were fed either a C or a HF diet after weaning, to generate four offspring groups; HF/HF, HF/C, C/HF, C/C. At 15 weeks of age, liver histology was normal in both the C/C and HF/C offspring. Kleiner scoring showed that although the C/HF offspring developed nonalcoholic fatty liver, the HF/HF offspring developed NASH. At 30 weeks, histological analysis and Kleiner scoring showed that both the HF/C and C/HF groups had NAFLD, whereas the HF/HF had a more severe form of NASH. Therefore, exposure to a HF diet in utero and during lactation contributes toward NAFLD progression. We investigated the mechanisms by which this developmental priming is mediated. At 15 weeks of age, hepatic mitochondrial electron transport chain (ETC) enzyme complex activity (I, II/III, and IV) was reduced in both groups of offspring from HF-fed mothers (HF/C and HF/HF). In addition, measurement of hepatic gene expression indicated that lipogenesis, oxidative stress, and inflammatory pathways were up-regulated in the 15-week-old HF/C and HF/HF offspring. CONCLUSION: Maternal fat intake contributes toward the NAFLD progression in adult offspring, which is mediated through impaired hepatic mitochondrial metabolism and up-regulated hepatic lipogenesis.


Assuntos
Gorduras na Dieta/administração & dosagem , Fígado Gorduroso/etiologia , Lipogênese , Fenômenos Fisiológicos da Nutrição Materna , Mitocôndrias Hepáticas/metabolismo , Animais , Modelos Animais de Doenças , Transporte de Elétrons , Feminino , Regulação da Expressão Gênica , Receptores de Hialuronatos/genética , Camundongos , Camundongos Endogâmicos C57BL , Estresse Oxidativo , Reação em Cadeia da Polimerase , Gravidez
7.
Am J Physiol Regul Integr Comp Physiol ; 297(3): R675-81, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19535678

RESUMO

We recently reported insulin resistance in adult offspring of obese C57BL/6J mice. We have now evaluated whether parameters of skeletal muscle structure and function may play a role in insulin resistance in this model of developmental programming. Obesity was induced in female mice by feeding a highly palatable sugar and fat-rich diet for 6 wk prior to pregnancy, and during pregnancy and lactation. Offspring of obese dams were weaned onto standard laboratory chow. At 3 mo of age, skeletal muscle insulin signaling protein expression, mitochondrial electron transport chain activity (ETC), muscle fiber type, fiber density, and fiber cross-sectional area were compared with that of offspring of control dams weaned onto the chow diet. Female offspring of obese dams demonstrated decreased skeletal muscle expression of p110beta, the catalytic subunit of PI3K (P < 0.01), as well as reduced Akt phosphorylation at Serine residue 473 compared with control offspring. Male offspring of obese dams demonstrated increased skeletal muscle Akt2 and PKCzeta expression (P < 0.01; P < 0.001, respectively). A decrease in mitochondrial-linked complex II-III was observed in male offspring of obese dams (P < 0.01), which was unrelated to CoQ deficiency. This was not observed in females. There were no differences in muscle fiber density between offspring of obese dams and control offspring in either sex. Sex-related alterations in key insulin-signaling proteins and in mitochondrial ETC may contribute to a state of insulin resistance in offspring of obese mice.


Assuntos
Complexo III da Cadeia de Transporte de Elétrons/metabolismo , Complexo II de Transporte de Elétrons/metabolismo , Resistência à Insulina , Insulina/metabolismo , Mitocôndrias Musculares/metabolismo , Obesidade/metabolismo , Músculo Quadríceps/metabolismo , Transdução de Sinais , Fenômenos Fisiológicos da Nutrição Animal , Animais , Peso Corporal , Classe I de Fosfatidilinositol 3-Quinases , Modelos Animais de Doenças , Feminino , Transportador de Glucose Tipo 4/metabolismo , Proteínas Substratos do Receptor de Insulina/metabolismo , Masculino , Fenômenos Fisiológicos da Nutrição Materna , Camundongos , Camundongos Endogâmicos C57BL , Mitocôndrias Musculares/enzimologia , Fibras Musculares Esqueléticas/metabolismo , Obesidade/patologia , Obesidade/fisiopatologia , Fosfatidilinositol 3-Quinases/metabolismo , Fosforilação , Gravidez , Efeitos Tardios da Exposição Pré-Natal , Proteína Quinase C/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Músculo Quadríceps/enzimologia , Músculo Quadríceps/patologia , Receptor de Insulina/metabolismo , Fatores Sexuais , Ubiquinona/metabolismo
8.
Biochim Biophys Acta ; 1783(10): 1935-42, 2008 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-18585410

RESUMO

Pluripotent mouse embryonic stem (mES) cells derived from the blastocyst of the preimplantation embryo can be induced to differentiate in vitro along different cell lineages. However the molecular and cellular factors that signal and/or determine the expression of key genes, and the localisation of the encoded proteins, during the differentiation events are poorly understood. One common mechanism by which proteins can be targeted to specific regions of the cell is through the asymmetric localisation of mRNAs and Staufen, a double-stranded RNA binding protein, is known to play a direct role in mRNA transport and localisation. The aims of the present study were to describe the expression of Staufen in preimplantation embryos and mES cells and to use RNA interference (RNAi) to investigate the roles of Staufen1 in mES cell lineage differentiation. Western blotting and immunocytochemistry demonstrated that Staufen is present in the preimplantation mouse embryo, pluripotent mES cells and mES cells stimulated to differentiate into embryoid bodies, but the Staufen staining patterns did not support asymmetric distribution of the protein. Knockdown of Staufen1 gene expression in differentiating mES cells reduced the synthesis of lineage-specific markers including Brachyury, alpha-fetoprotein (AFP), PAX-6, and Vasa. There was however no significant change in either the gene expression of Nanog and Oct4, or in the synthesis of SSEA-1, all of which are key markers of pluripotency. These data indicate that inhibition of Staufen1 gene expression by RNAi affects an early step in mES cell differentiation and suggest a key role for Staufen in the cell lineage differentiation of mES cells.


Assuntos
Blastocisto/citologia , Blastocisto/metabolismo , Diferenciação Celular , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/metabolismo , Proteínas de Ligação a RNA/metabolismo , Animais , Biomarcadores , Proliferação de Células , Células Cultivadas , Regulação da Expressão Gênica no Desenvolvimento , Camundongos , RNA Interferente Pequeno/genética , Proteínas de Ligação a RNA/genética
9.
Hypertension ; 51(2): 383-92, 2008 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-18086952

RESUMO

Maternal obesity is increasingly prevalent and may affect the long-term health of the child. We investigated the effects of maternal diet-induced obesity in mice on offspring metabolic and cardiovascular function. Female C57BL/6J mice were fed either a standard chow (3% fat, 7% sugar) or a palatable obesogenic diet (16% fat, 33% sugar) for 6 weeks before mating and throughout pregnancy and lactation. Offspring of control (OC) and obese dams (OO) were weaned onto standard chow and studied at 3 and 6 months of age. OO were hyperphagic from 4 to 6 weeks of age compared with OC and at 3 months locomotor activity was reduced and adiposity increased (abdominal fat pad mass; P<0.01). OO were heavier than OC at 6 months (body weight, P<0.05). OO abdominal obesity was associated with adipocyte hypertrophy and altered mRNA expression of beta-adrenoceptor 2 and 3, 11 beta HSD-1, and PPAR-gamma 2. OO showed resistance artery endothelial dysfunction at 3 months, and were hypertensive, as assessed by radiotelemetry (nighttime systolic blood pressure at 6 months [mm Hg] mean+/-SEM, male OO, 134+/-1 versus OC, 124+/-2, n=8, P<0.05; female OO, 137+/-2 versus OC, 122+/-4, n=8, P<0.01). OO skeletal muscle mass (tibialis anterior) was significantly reduced (P<0.01) OO fasting insulin was raised at 3 months and by 6 months fasting plasma glucose was elevated. Exposure to the influences of maternal obesity in the developing mouse led to adult offspring adiposity and cardiovascular and metabolic dysfunction. Developmentally programmed hyperphagia, physical inactivity, and altered adipocyte metabolism may play a mechanistic role.


Assuntos
Adiposidade , Dieta , Hiperfagia/etiologia , Hipertensão/etiologia , Resistência à Insulina , Obesidade/etiologia , Complicações na Gravidez , Efeitos Tardios da Exposição Pré-Natal , Adipócitos/patologia , Adiposidade/genética , Animais , Artérias/fisiopatologia , Pressão Sanguínea , Capilares/patologia , Tamanho Celular , Feminino , Expressão Gênica , Teste de Tolerância a Glucose , Frequência Cardíaca , Insulina/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Obesidade/complicações , Obesidade/patologia , Obesidade/fisiopatologia , Pâncreas/metabolismo , Gravidez , Resistência Vascular
10.
Mech Ageing Dev ; 128(11-12): 681-7, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-18035399

RESUMO

We have recently reported that maternal dietary imbalance during pregnancy and lactation can reduce the lifespan of offspring. Rats that were growth restricted in utero by maternal protein restriction and underwent rapid weight gain when suckled by control fed dams died earlier than animals whose mothers were fed a control diet throughout pregnancy and lactation. We demonstrate here that mitochondrial abnormalities and DNA damage occur in the kidney of offspring who die prematurely. We have established by direct measurement and by in vitro supplementation that mitochondrial abnormalities occur because of a functional deficit of the mitochondrial cofactor coenzyme Q9 (CoQ9). These data provide molecular insight into the association between maternal nutrition and determination of offspring lifespan, and identify, a potential dietary intervention to prevent detrimental consequences of imbalanced maternal nutrition.


Assuntos
Dieta com Restrição de Proteínas , Retardo do Crescimento Fetal/fisiopatologia , Rim/metabolismo , Longevidade , Efeitos Tardios da Exposição Pré-Natal , Fenômenos Fisiológicos da Nutrição Pré-Natal , Ubiquinona/deficiência , Aumento de Peso , Animais , Animais Recém-Nascidos , Coenzimas/deficiência , Dano ao DNA , Modelos Animais de Doenças , Complexo de Proteínas da Cadeia de Transporte de Elétrons/metabolismo , Feminino , Retardo do Crescimento Fetal/genética , Retardo do Crescimento Fetal/metabolismo , Rim/enzimologia , Lactação , Masculino , Mitocôndrias/metabolismo , Gravidez , Ratos , Ratos Wistar , Telômero/metabolismo , Fatores de Tempo , Ubiquinona/análogos & derivados
11.
Mol Reprod Dev ; 71(4): 399-404, 2005 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-15880683

RESUMO

T-box genes are a highly conserved family of genes encoding transcription factors, which share a conserved DNA binding domain (the T-box). Appropriate temporal and spatial expression of this gene family is critical for gastrulation and organogenesis in a number of species. The T-box containing gene Eomesodermin was first identified in Xenopus, where it plays a critical role in mesoderm formation. In situ analyses in mice have described the expression patterns of the mouse ortholog of this gene mEomesodermin (mEomes) at the time of implantation and during fetal development. Additional studies involving the disruption of the mEomes gene, have demonstrated an additional role for mEomes in trophoblast formation. However, these analyses did not address the possibility that maternally encoded or pre-blastocyst zygotic transcription of mEomes may also contribute to embryonic development. We show here that mEomes mRNA is present prior to blastocyst formation, and that the protein product of mEomes is associated with nuclear DNA during oocyte development and persistently localizes within all nuclei of the preimplantation embryo until the early blastocyst stage. mEomes protein is associated with the meiotic spindle in the unfertilized egg and with the mitotic spindle at each cell division. Our results are consistent with mEomesodermin having a role in early preimplantation development and inner cell mass formation in addition to its function in the trophoblast lineage.


Assuntos
Blastocisto/metabolismo , Oócitos/metabolismo , Proteínas com Domínio T/metabolismo , Animais , Western Blotting , Reações Cruzadas , Feminino , Camundongos , Microscopia Confocal , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Proteínas com Domínio T/genética , Proteínas com Domínio T/imunologia
12.
FEBS Lett ; 579(10): 2226-30, 2005 Apr 11.
Artigo em Inglês | MEDLINE | ID: mdl-15811346

RESUMO

mRNA localisation, as a mechanism for directing localised protein synthesis, plays a vital role in the functioning of certain cells, such as neurons and oocytes. Potentially this mechanism may also occur in polarised intestinal epithelial cells. Here we show that Staufen1(55), a protein involved in mRNA localisation and transport, is asymmetrically distributed in differentiated Caco-2 intestinal epithelial cells and partly co-localised with calnexin, a marker of the endoplasmic reticulum. The localisation to the apical region of the cell indicates that Staufen may be involved in localisation of transcripts to this domain.


Assuntos
Mucosa Intestinal/metabolismo , Proteínas de Ligação a RNA/metabolismo , Células CACO-2 , Diferenciação Celular , Proteínas do Citoesqueleto , Células Epiteliais/citologia , Células Epiteliais/metabolismo , Humanos , Mucosa Intestinal/citologia
13.
Am J Physiol Regul Integr Comp Physiol ; 288(1): R134-9, 2005 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-15388492

RESUMO

We previously reported that prenatal and suckling exposure to a maternal diet rich in animal fat leads to cardiovascular dysfunction in young adult rat offspring with subsequent development of dyslipidemia and hyperglycemia. We have further investigated glucose homeostasis in adult female offspring by euglycemic-hyperinsulinemic clamp and by dynamic assessment of glucose-stimulated insulin secretion in isolated, perifused pancreatic islet cells. Additionally, given the link between reduced mitochondrial DNA (mtDNA) content and the development of type 2 diabetes mellitus, we have measured mtDNA in organs from young adult animals. Sprague-Dawley rats were fed a diet rich in animal fat or normal chow throughout pregnancy and weaning. Infusion of insulin (5 mU.kg(-1).min(-1)) resulted in a higher steady-state plasma insulin concentration in 1-year-old offspring of fat-fed dams (OHF, n = 4) vs. offspring of control dams (OC, n = 4, P < 0.01). Glucose-stimulated insulin secretion in isolated islets from 9-mo-old OHF was significantly reduced compared with OC (n = 4, P < 0.05). Transmission electron micrography showed altered insulin secretory granule morphology in OHF pancreatic beta-cells. Kidney mtDNA was reduced in 3-mo-old OHF [16S-to-18S gene ratio: OC (n = 10) 1.05 +/- 0.19 vs. OHF (n = 10) 0.66 +/- 0.06, P < 0.05]. At 6 mo, gene chip microarray of OHF aorta showed reduced expression of the mitochondrial genome. Prenatal and suckling exposure to a diet rich in animal fat leads to whole body insulin resistance and pancreatic beta-cell dysfunction in adulthood, which is preceded by reduced tissue mtDNA content and altered mitochondrial gene expression.


Assuntos
Glicemia/fisiologia , Gorduras na Dieta/farmacologia , Mitocôndrias/metabolismo , Efeitos Tardios da Exposição Pré-Natal , Fenômenos Fisiológicos da Nutrição Pré-Natal/fisiologia , Tecido Adiposo/crescimento & desenvolvimento , Animais , Peso Corporal , Feminino , Expressão Gênica/fisiologia , Homeostase , Insulina/sangue , Resistência à Insulina/fisiologia , Ilhotas Pancreáticas/fisiologia , Leptina/sangue , Lipídeos/sangue , Síndrome Metabólica/embriologia , Gravidez , Ratos , Ratos Sprague-Dawley
14.
Reprod Biomed Online ; 9(4): 418-24, 2004 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-15511342

RESUMO

There is increasing evidence in humans that abnormal mitochondrial DNA (mtDNA) is associated with common degenerative disorders of the twenty-first century. MtDNA is exclusively female in origin and abnormalities in mtDNA can either be inherited, or generated de novo by adverse environmental factors that disturb mitochondrial DNA synthesis or destabilize mtDNA. The preimplantation period of development in mammals was thought to be relatively immune from environmentally induced changes to mtDNA, since no replication of mtDNA was thought to occur at this stage. This study demonstrates that there is a very short period of mtDNA synthesis immediately after fertilization, which can be affected by environmental stress. Adverse culture conditions during this phase of development could therefore alter the mitochondrial genome, with possible long-term consequences for the health of the offspring. The findings have relevance for all assisted reproduction programmes and for the rapidly emerging field of stem cell technologies.


Assuntos
DNA Mitocondrial/genética , DNA Mitocondrial/metabolismo , Desenvolvimento Embrionário/genética , Desenvolvimento Embrionário/fisiologia , Animais , Sequência de Bases , Blastocisto/metabolismo , Fase de Clivagem do Zigoto/metabolismo , Replicação do DNA , Feminino , Dosagem de Genes , Humanos , Camundongos , Gravidez , Técnicas de Cultura de Tecidos , Zigoto/metabolismo
15.
Cancer Epidemiol Biomarkers Prev ; 13(9): 1436-43, 2004 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-15342443

RESUMO

OBJECTIVE: Homozygosity for variants of the methylenetetrahydrofolate reductase (MTHFR) gene is associated with decreased risk for colorectal cancer. We have investigated the relationships between two variants of the MTHFR gene (C677T and A1298C) and blood folate, homocysteine, and genomic stability (strand breakage, misincorporated uracil, and global cytosine methylation in lymphocytes) in a study of 199 subjects. RESULTS: The frequencies of homozygosity for the C677T and A1298C variants of the MTHFR gene were 12.6% and 14.6%, respectively. Plasma homocysteine, folate, vitamin B12, 5-methyltetrahydrofolate, and RBC folate were determined in the C677T genotypes. Plasma folate was significantly lower (P < 0.001) in the homozygous variants (6.7 +/- 0.6 ng/mL) compared with wild-types (8.8 +/- 0.4 ng/mL) and heterozygotes (9.1 +/- 0.5 ng/mL). Homocysteine was significantly higher (P < 0.05) in homozygous variants (13.2 +/- 1.1 micromol/L) compared with homozygous subjects (10.9 +/- 0.4 micromol/L). Homozygous variants had significantly lower (P < 0.05) RBC folate (84.7 +/- 6.3 ng/mL) compared with wild-types (112.2 +/- 5.2 ng/mL) and heterozygous individuals (125.1 +/- 6.6 ng/mL). No significant difference in RBC folate was observed between wild-types and heterozygotes. The A1298C variant did not influence plasma homocysteine, folate, 5-methyltetrahydrofolate, vitamin B12, or RBC folate. Lymphocyte DNA stability biomarkers (strand breaks, misincorporated uracil, and global DNA methylation) were similar for all MTHFR C677T or A1298C variants. CONCLUSION: Data from this study do not support the hypothesis that polymorphisms in the MTHFR gene increase DNA stability by sequestering 5,10-methylenetetrahydrofolate for thymidine synthesis and reducing uracil misincorporation into DNA.


Assuntos
Neoplasias Colorretais/genética , Dano ao DNA/genética , Metilação de DNA , Ácido Fólico/sangue , Variação Genética/genética , Genótipo , Metilenotetra-Hidrofolato Redutase (NADPH2)/genética , Uracila/metabolismo , Adulto , Idoso , Idoso de 80 Anos ou mais , Estudos de Casos e Controles , Neoplasias Colorretais/enzimologia , Neoplasias Colorretais/epidemiologia , Eritrócitos/enzimologia , Feminino , Triagem de Portadores Genéticos , Homocisteína/sangue , Homozigoto , Humanos , Isoenzimas/genética , Linfócitos/enzimologia , Masculino , Pessoa de Meia-Idade , Polimorfismo Genético/genética , Risco , Escócia , Tetra-Hidrofolatos/sangue , Vitamina B 12/sangue
16.
Semin Cell Dev Biol ; 15(5): 583-97, 2004 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-15271304

RESUMO

The first developmental lineage allocation during the generation of the mouse blastocyst is to outer trophoblast or to inner pluriblast (inner cell mass; ICM) cells. This allocation seems to be initiated at the 8-cell stage, when blastomeres polarise. Polarisation is followed by differentiative divisions at the subsequent two cleavage divisions to generate polar outer and non-polar inner 16- and 32-cells. The key events in polarisation are regulated post-translationally through a cell contact-mediated pathway, which imposes a heritable determinant-like organisation on the blastomere cortex. Two proteins in particular, E-cadherin and ezrin, are intimately involved in the generation and stabilisation of developmentally significant information. Transcriptional differences between lineages appear to follow and may coincide with the lineage commitment of cells.


Assuntos
Padronização Corporal/fisiologia , Linhagem da Célula/fisiologia , Animais , Blastômeros/fisiologia , Caderinas , Proteínas do Citoesqueleto , Feminino , Camundongos , Óvulo/fisiologia , Fosfoproteínas/fisiologia , Trofoblastos/fisiologia
17.
Br J Nutr ; 88(5): 471-7, 2002 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-12425727

RESUMO

Maternal malnutrition can lead to fetal abnormalities and increase susceptibility to disease in later life. Rat models have been developed to study the physiology and metabolism underlying this phenomenon. One particular model of 50 % protein restriction during pregnancy, the low-protein diet (LPD) supplemented with methionine, has been developed to investigate the underlying mechanisms. Recent studies have shown that rats fed a LPD during only the first 4 d of pregnancy produce offspring that develop hypertension. These results suggest that the very earliest stages of embryo development are susceptible to diet-induced heritable changes. We demonstrate a marked elevation of maternal serum homocysteine (hcy) concentrations during the initial phases of pregnancy in both rats and mice fed an LPD. Fetal growth and many of the circulating amino acids are similarly perturbed in both rats and mice fed the LPD during pregnancy, indicating that the response to the LPD diet is similar in rats and mice. These findings allow us to exploit the advantages of the mouse experimental system in future analyses aimed at understanding the molecular basis of fetal programming. Our present findings are discussed with particular reference to mechanisms which may initiate fetal programming, and to the feasibility of dietary interventions aimed at reducing early pregnancy loss and pre-eclampsia in man.


Assuntos
Blastocisto/metabolismo , Dieta com Restrição de Proteínas , Homocisteína/sangue , Modelos Animais , Fenômenos Fisiológicos da Nutrição Pré-Natal , Aborto Espontâneo/etiologia , Aminoácidos/sangue , Animais , Suscetibilidade a Doenças , Desenvolvimento Embrionário e Fetal , Feminino , Idade Gestacional , Metionina/administração & dosagem , Camundongos , Camundongos Endogâmicos , Pré-Eclâmpsia/etiologia , Gravidez , Ratos , Ratos Endogâmicos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA