Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 45
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Biochem Biophys Res Commun ; 716: 149998, 2024 07 05.
Artigo em Inglês | MEDLINE | ID: mdl-38692012

RESUMO

The equilibrium between the hypertrophic growth of existing adipocytes and adipogenesis is vital in managing metabolic stability in white adipocytes when faced with overnutrition. Adipogenesis has been established as a key player in combating metabolic irregularities caused by various factors. However, the benefits of increasing adipogenesis-mediated white adipose tissue (WAT) expansion for metabolic health regulation remain uncertain. Our findings reveal an increase in Impdh2 expression during the adipogenesis phase, both in vivo and in vitro. Xmp enhances adipogenic potential by fostering mitotic clonal expansion (MCE). The conditional knockout of Impdh2 in adipocyte progenitor cells(APCs) in adult and aged mice effectively curbs white adipose tissue expansion, ameliorates glucose tolerance, and augments energy expenditure under high-fat diet (HFD). However, no significant difference is observed under normal chow diet (NCD). Concurrently, the knockout of Impdh2 in APCs significantly reduces the count of new adipocytes induced by HFD, without affecting adipocyte size. Mechanistically, Impdh2 regulates the proliferation of APCs during the MCE phase via Xmp. Exogenous Xmp can significantly offset the reduction in adipogenic abilities of APCs due to Impdh2 deficiency. In summary, we discovered that adipogenesis-mediated WAT expansion, induced by overnutrition, also contributes to metabolic abnormalities. Moreover, the pivotal role of Impdh2 in regulating adipogenesis in APCs offers a novel therapeutic approach to combat obesity.


Assuntos
Adipócitos , Adipogenia , Tecido Adiposo Branco , Dieta Hiperlipídica , IMP Desidrogenase , Hipernutrição , Animais , Masculino , Camundongos , Adipócitos/metabolismo , Adipogenia/genética , Tecido Adiposo Branco/metabolismo , Proliferação de Células , Metabolismo Energético/genética , Deleção de Genes , Camundongos Endogâmicos C57BL , Camundongos Knockout , Hipernutrição/metabolismo , Hipernutrição/genética , Células-Tronco/metabolismo , Células-Tronco/citologia , Células-Tronco/patologia , IMP Desidrogenase/genética , IMP Desidrogenase/metabolismo
2.
Am J Physiol Endocrinol Metab ; 321(5): E702-E713, 2021 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-34632797

RESUMO

In chronic obesity, activated adipose tissue proinflammatory cascades are tightly linked to metabolic dysfunction. Yet, close temporal analyses of the responses to obesogenic environment such as high-fat feeding (HFF) in susceptible mouse strains question the causal relationship between inflammation and metabolic dysfunction, and/or raises the possibility that certain inflammatory cascades play adaptive/homeostatic, rather than pathogenic roles. Here, we hypothesized that CTRP6, a C1QTNF family member, may constitute an early responder to acute nutritional changes in adipose tissue, with potential physiological roles. Both 3-days high-fat feeding (3dHFF) and acute obesity reversal [2-wk switch to low-fat diet after 8-wk HFF (8wHFF)] already induced marked changes in whole body fuel utilization. Although adipose tissue expression of classical proinflammatory cytokines (Tnf-α, Ccl2, and Il1b) exhibited no, or only minor, change, C1qtnf6 uniquely increased, and decreased, in response to 3dHFF and acute obesity reversal, respectively. CTRP6 knockout (KO) mouse embryonic fibroblasts (MEFs) exhibited increased adipogenic gene expression (Pparg, Fabp4, and Adipoq) and markedly reduced inflammatory genes (Tnf-α, Ccl2, and Il6) compared with wild-type MEFs, and recombinant CTRP6 induced the opposite gene expression signature, as assessed by RNA sequencing. Consistently, 3dHFF of CTRP6-KO mice induced a greater whole body and adipose tissue weight gain compared with wild-type littermates. Collectively, we propose CTRP6 as a gene that rapidly responds to acute changes in caloric intake, acting in acute overnutrition to induce a "physiological inflammatory response" that limits adipose tissue expansion.NEW & NOTEWORTHY CTRP6 (C1qTNF6), a member of adiponectin gene family, regulates inflammation and metabolism in established obesity. Here, short-term high-fat feeding in mice is shown to increase adipose tissue expression of CTRP6 before changes in the expression of classical inflammatory genes occur. Conversely, CTRP6 expression in adipose tissue decreases early in the course of obesity reversal. Gain- and loss-of-function models suggest CTRP6 as a positive regulator of inflammatory cascades, and a negative regulator of adipogenesis and adipose tissue expansion.


Assuntos
Adipocinas/fisiologia , Tecido Adiposo/patologia , Inflamação/genética , Fenômenos Fisiológicos da Nutrição/genética , Adipogenia/genética , Adipocinas/genética , Tecido Adiposo/metabolismo , Animais , Células Cultivadas , Dieta Hiperlipídica , Embrião de Mamíferos , Feminino , Células HEK293 , Humanos , Inflamação/metabolismo , Inflamação/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Tamanho do Órgão/genética , Hipernutrição/genética , Hipernutrição/metabolismo , Hipernutrição/patologia , Gravidez
3.
IUBMB Life ; 73(10): 1210-1221, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34184397

RESUMO

Pathological states in the early life environment of mammalian offspring, including maternal obesity and intrauterine overnutrition, can induce obesity and metabolic disorder later in life. Leptin resistance caused by upregulation of Socs3 in the hypothalamus of offspring was believed to be the main mechanism of this effect. In this study, obese mother (OM) and lean mother (LM) models were generated by feeding C57BL/6N female mice a high-fat diet or standard lean diet, respectively. Additionally, an obese mother with intervention (OMI) model was generated by injecting the high-fat diet group with Socs3-shRNA lentivirus during early pregnancy. The offspring of the groups was correspondingly named OM-F1 , LM-F1 , and OMI-F1 , representing progeny mouse models of different early life environments. The offspring were fed a high-fat diet to test their propensity for obesity. The body weight, food intake and fat accumulation were higher, while glucose intolerance and insulin resistance were worse in the OM-F1 group than LM-F1 group. By contrast, the obesity phenotype, hyperphagia and metabolic disorder were alleviated in the OMI-F1 group compared with the OM-F1 group. The mechanism was identified that downregulation of hypothalamic SOCS3 resulted in an increased level of p-STAT3 and p-JAK2, which ameliorated the leptin resistance and restored the lean expression of appetite regulatory genes (Pomc and Agrp) in hypothalamus of OMI-F1 group. Taken together, these results indicate that reducing maternal Socs3 expression during pregnancy can attenuate obesity caused by the early life environment in mice, which may inspire therapies that enable obese mothers to bear metabolically healthy children.


Assuntos
Obesidade Materna/genética , Hipernutrição/genética , Proteína 3 Supressora da Sinalização de Citocinas/genética , Tecido Adiposo , Animais , Animais Recém-Nascidos , Apetite/genética , Peso Corporal/genética , Modelos Animais de Doenças , Regulação para Baixo , Ingestão de Alimentos , Feminino , Técnicas de Silenciamento de Genes , Fígado/metabolismo , Masculino , Camundongos Endogâmicos C57BL , Hipernutrição/complicações , Gravidez
4.
Int J Mol Sci ; 22(4)2021 Feb 20.
Artigo em Inglês | MEDLINE | ID: mdl-33672754

RESUMO

A high-fat, Western-style diet is an important predisposing factor for the onset of type 2 diabetes and obesity. It causes changes in gut microbial profile, reduction of microbial diversity, and the impairment of the intestinal barrier, leading to increased serum lipopolysaccharide (endotoxin) levels. Elevated lipopolysaccharide (LPS) induces acetyltransferase P300 both in the nucleus and cytoplasm of liver hepatocytes through the activation of the IRE1-XBP1 pathway in the endoplasmic reticulum stress. In the nucleus, induced P300 acetylates CRTC2 to increase CRTC2 abundance and drives Foxo1 gene expression, resulting in increased expression of the rate-limiting gluconeogenic gene G6pc and Pck1 and abnormal liver glucose production. Furthermore, abnormal cytoplasm-appearing P300 acetylates IRS1 and IRS2 to disrupt insulin signaling, leading to the prevention of nuclear exclusion and degradation of FOXO1 proteins to further exacerbate the expression of G6pc and Pck1 genes and liver glucose production. Inhibition of P300 acetyltransferase activity by chemical inhibitors improved insulin signaling and alleviated hyperglycemia in obese mice. Thus, P300 acetyltransferase activity appears to be a therapeutic target for the treatment of type 2 diabetes and obesity.


Assuntos
Microbioma Gastrointestinal/genética , Regulação da Expressão Gênica , Gluconeogênese/genética , Insulina/metabolismo , Hipernutrição/genética , Hipernutrição/microbiologia , Transdução de Sinais , Animais , Humanos , Hipernutrição/sangue
5.
Mamm Genome ; 31(5-6): 119-133, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-32350605

RESUMO

Nutritional constraints including not only caloric restriction or protein deficiency, but also energy-dense diets affect metabolic health and frequently lead to obesity and insulin resistance, as well as glucose intolerance and type 2 diabetes. The effects of these environmental factors are often mediated via epigenetic modifiers that target the expression of metabolic genes. More recently, it was discovered that such parentally acquired metabolic changes can alter the metabolic health of the filial and grand-filial generations. In mammals, this epigenetic inheritance can either follow an intergenerational or transgenerational mode of inheritance. In the case of intergenerational inheritance, epimutations established in gametes persist through the first round of epigenetic reprogramming occurring during preimplantation development. For transgenerational inheritance, epimutations persist additionally throughout the reprogramming that occurs during germ cell development later in embryogenesis. Differentially expressed transcripts, genomic cytosine methylations, and several chemical modifications of histones are prime candidates for tangible marks which may serve as epimutations in inter- and transgenerational inheritance and which are currently being investigated experimentally. We review, here, the current literature in support of epigenetic inheritance of metabolic traits caused by nutritional constraints and potential mechanisms in man and in rodent model systems.


Assuntos
Diabetes Mellitus Tipo 2/genética , Intolerância à Glucose/genética , Padrões de Herança , Desnutrição/genética , Obesidade/genética , Hipernutrição/genética , Animais , Metilação de DNA , Diabetes Mellitus Tipo 2/etiologia , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/patologia , Desenvolvimento Embrionário/genética , Disruptores Endócrinos/administração & dosagem , Feminino , Interação Gene-Ambiente , Intolerância à Glucose/metabolismo , Intolerância à Glucose/patologia , Histonas/genética , Histonas/metabolismo , Humanos , Resistência à Insulina/genética , Masculino , Desnutrição/complicações , Desnutrição/metabolismo , Desnutrição/patologia , Obesidade/etiologia , Obesidade/metabolismo , Obesidade/patologia , Hipernutrição/complicações , Hipernutrição/metabolismo , Hipernutrição/patologia , RNA Mensageiro/genética , RNA Mensageiro/metabolismo
6.
Hepatology ; 69(3): 1122-1134, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30230575

RESUMO

Regulation of the protein stability of epigenetic regulators remains ill-defined despite its potential applicability in epigenetic therapies. The histone H3-lysine 4-methyltransferase MLL4 is an epigenetic transcriptional coactivator that directs overnutrition-induced obesity and fatty liver formation, and Mll4+/- mice are resistant to both. Here we show that the E3 ubiquitin ligase UBE3A targets MLL4 for degradation, thereby suppressing high-fat diet (HFD)-induced expression of the hepatic steatosis target genes of MLL4. In contrast to Mll4+/- mice, Ube3a+/- mice are hypersensitive to HFD-induced obesity and fatty liver development. Ube3a+/-;Mll4+/- mice lose this hypersensitivity, supporting roles of increased MLL4 levels in both phenotypes of Ube3a+/- mice. Correspondingly, our comparative studies with wild-type, Ube3a+/- and Ube3a-/- and UBE3A-overexpressing transgenic mouse livers demonstrate an inverse correlation of UBE3A protein levels with MLL4 protein levels, expression of the steatosis target genes of MLL4, and their decoration by H3-lysine 4-monomethylation, a surrogate marker for the epigenetic action of MLL4. Conclusion: UBE3A indirectly exerts an epigenetic regulation of obesity and steatosis by degrading MLL4. This UBE3A-MLL4 regulatory axis provides a potential therapeutic venue for treating various MLL4-directed pathogeneses, including obesity and hepatic steatosis.


Assuntos
Fígado Gorduroso/genética , Regulação da Expressão Gênica/fisiologia , Histona-Lisina N-Metiltransferase/metabolismo , Hipernutrição/genética , Ubiquitina-Proteína Ligases/fisiologia , Animais , Feminino , Masculino , Camundongos
7.
Int J Obes (Lond) ; 42(8): 1431-1444, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-29777232

RESUMO

BACKGROUND AND OBJECTIVE: Maternal overnutrition has been implicated in affecting the offspring by programming metabolic disorders such as obesity and diabetes, by mechanisms that are not clearly understood. This study aimed to determine the long-term impact of maternal high-fat (HF) diet feeding on epigenetic changes in the offspring's hypothalamic Pomc gene, coding a key factor in the control of energy balance. Further, it aimed to study the additional effects of postnatal overnutrition on epigenetic programming by maternal nutrition. METHODS: Eight-week-old female Sprague-Dawley rats were fed HF diet or low-fat (LF) diet for 6 weeks before mating, and throughout gestation and lactation. At postnatal day 21, samples were collected from a third offspring and the remainder were weaned onto LF diet for 5 weeks, after which they were either fed LF or HF diet for 12 weeks, resulting in four groups of offspring differing by their maternal and postweaning diet. RESULTS: With maternal HF diet, offspring at weaning had rapid early weight gain, increased adiposity, and hyperleptinemia. The programmed adult offspring, subsequently fed LF diet, retained the increased body weight. Maternal HF diet combined with offspring HF diet caused more pronounced hyperphagia, fat mass, and insulin resistance. The ARC Pomc gene from programmed offspring at weaning showed hypermethylation in the enhancer (nPE1 and nPE2) regions and in the promoter sequence mediating leptin effects. Interestingly, hypermethylation at the Pomc promoter but not at the enhancer region persisted long term into adulthood in the programmed offspring. However, there were no additive effects on methylation levels in the regulatory regions of Pomc in programmed offspring fed a HF diet. CONCLUSION: Maternal overnutrition programs long-term epigenetic alterations in the offspring's hypothalamic Pomc promoter. This predisposes the offspring to metabolic disorders later in life.


Assuntos
Epigênese Genética/genética , Hipotálamo/metabolismo , Fenômenos Fisiológicos da Nutrição Materna/genética , Hipernutrição/genética , Efeitos Tardios da Exposição Pré-Natal/genética , Pró-Opiomelanocortina/genética , Sequências Reguladoras de Ácido Nucleico/genética , Animais , Metilação de DNA , Modelos Animais de Doenças , Feminino , Hipotálamo/química , Obesidade/genética , Obesidade/metabolismo , Hipernutrição/metabolismo , Hipernutrição/fisiopatologia , Gravidez , Pró-Opiomelanocortina/metabolismo , Ratos , Ratos Sprague-Dawley
8.
Rheumatology (Oxford) ; 57(10): 1769-1776, 2018 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-29939348

RESUMO

Objective: Gout is the most common form of inflammatory arthritis and is caused by hyperuricaemia. Some studies have found a reduction in plasma urate with vitamin C supplementation. We tested the hypothesis that high plasma vitamin C is causally associated with low plasma urate and low risk of hyperuricaemia, using a Mendelian randomization approach. Methods: We measured plasma urate and genotyped for the SLC23A1 rs33972313 vitamin C variant in 106 147 individuals from the Copenhagen General Population Study, of which 24 099 had hyperuricaemia. We measured plasma vitamin C in 9234 individuals and genotyped for the SLC2A9 rs7442295 urate variant in 102 345 individuals. Results: Each 10 µmol/l higher plasma vitamin C was associated with a -2.3(95%CI: -0.69 to -3.9) µmol/l lower plasma urate after multivariable adjustments. The SLC23A1 rs33972313 GG genotype was associated with a 9% (5.6%, 11.9%) higher plasma vitamin C compared with AA and AG combined but was not associated with plasma urate (P = 0.31). Likewise, for each 10 µmol/l higher plasma vitamin C the odds ratios for hyperuricaemia were 0.92 (0.86, 0.98) observationally after multivariable adjustments, but 1.01 (0.84, 1.23) genetically. Conclusion: High plasma vitamin C was associated with low plasma urate and with low risk of hyperuricaemia. However, the SLC23A1 genetic variant causing lifelong high plasma vitamin C was not associated with plasma urate levels or with risk of hyperuricaemia. Thus, our data do not support a causal relationship between high plasma vitamin C and low plasma urate.


Assuntos
Ácido Ascórbico/sangue , Hiperuricemia/genética , Hipernutrição/genética , Ácido Úrico/sangue , Adulto , Idoso , Idoso de 80 Anos ou mais , Feminino , Variação Genética , Genótipo , Humanos , Hiperuricemia/sangue , Masculino , Análise da Randomização Mendeliana , Pessoa de Meia-Idade , Razão de Chances , Hipernutrição/sangue , Fatores de Risco , Adulto Jovem
9.
Nutr Metab Cardiovasc Dis ; 28(9): 944-951, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-29752038

RESUMO

BACKGROUND AND AIMS: The prevalence of obesity is increasing worldwide at an alarming rate. Altered early nutrition, in particular postnatal overfeeding (PNOF), is a risk factor for impaired cardiac function in adulthood. In the understanding of the initiation or progression of heart diseases, NLRP3 inflammasome and non-coding RNAs have been proposed as key players. In this context, the aim of this study was to decipher the role of NLRP3 inflammasome and its post transcriptional control by micro-RNAs in the regulation of cardiac metabolic function induced by PNOF in mice. METHODS AND RESULTS: Based on a model of mice exposed to PNOF through litter size reduction, we observed increased cardiac protein expression levels of NLRP3 and ETS-1 associated with alterations in insulin signaling. Additionally, miR-193b levels were down-regulated in the adult hearts of overfed animals. In a cardiomyocyte cell line, transfection with miR-193b induced down-regulation of ETS-1 and NLRP3 and improved insulin signaling. CONCLUSIONS: These findings suggest that the miR-193b could be involved in cardiac phenotypic changes observed in adulthood induced by PNOF likely through the regulation of ETS-1 and NLRP3 expression, and through this of insulin signaling.


Assuntos
Fenômenos Fisiológicos da Nutrição Animal , Cardiopatias/etiologia , Inflamassomos/metabolismo , Miocárdio/metabolismo , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Estado Nutricional , Hipernutrição/complicações , Animais , Animais Recém-Nascidos , Linhagem Celular , Modelos Animais de Doenças , Cardiopatias/genética , Cardiopatias/metabolismo , Cardiopatias/fisiopatologia , Insulina/metabolismo , Tamanho da Ninhada de Vivíparos , Camundongos Endogâmicos C57BL , MicroRNAs/genética , MicroRNAs/metabolismo , Hipernutrição/genética , Hipernutrição/metabolismo , Hipernutrição/fisiopatologia , Proteína Proto-Oncogênica c-ets-1/metabolismo , Ratos , Transdução de Sinais , Fatores de Tempo
10.
Nutr Metab Cardiovasc Dis ; 27(10): 930-937, 2017 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-28958692

RESUMO

BACKGROUND AND AIMS: The aim of this study was to analyze the effects of early overnutrition (EON) on the expression of the renin angiotensin aldosterone system (RAAS) components in renal cortex, renal arteries and renal perivascular adipose tissue (PVAT), as well as the vascular response of renal arteries to Angiotensin II (Ang II). METHODS AND RESULTS: On birth day litters were adjusted to twelve (L12-control) or three (L3-overfed) pups per mother. Half of the animals were sacrificed at weaning (21 days old) and the other half at 5 months of age. Ang II-induced vasoconstriction of renal artery segments increased in young overfed rats and decreased in adult overfed rats. EON decreased the gene expression of angiotensinogen (Agt), Ang II receptors AT1 and AT2 and eNOS in renal arteries of young rats, while it increased the mRNA levels of AT-2 and ET-1 in adult rats. In renal PVAT EON up-regulated the gene expression of COX-2 and TNF-α in young rats and the mRNA levels of renin receptor both in young and in adult rats. On the contrary, Ang II receptors mRNA levels were downregulated at both ages. Renal cortex of overfed rats showed increased gene expression of Agt in adult rats and of AT1 in young rats. However the mRNA levels of AT1 were decreased in the renal cortex of overfed adult rats. CONCLUSION: EON is associated with alterations in the vascular response of renal arteries to Ang II and changes in the gene expression of RAAS components in renal tissue.


Assuntos
Angiotensina II/farmacologia , Rim/irrigação sanguínea , Hipernutrição/metabolismo , Artéria Renal/efeitos dos fármacos , Sistema Renina-Angiotensina/efeitos dos fármacos , Vasoconstrição/efeitos dos fármacos , Vasoconstritores/farmacologia , Tecido Adiposo/efeitos dos fármacos , Tecido Adiposo/metabolismo , Tecido Adiposo/fisiopatologia , Fatores Etários , Fenômenos Fisiológicos da Nutrição Animal , Animais , Ciclo-Oxigenase 2/genética , Ciclo-Oxigenase 2/metabolismo , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Regulação da Expressão Gênica , Óxido Nítrico Sintase Tipo III/genética , Óxido Nítrico Sintase Tipo III/metabolismo , Estado Nutricional , Hipernutrição/genética , Hipernutrição/fisiopatologia , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Ratos Sprague-Dawley , Receptor Tipo 1 de Angiotensina/genética , Receptor Tipo 1 de Angiotensina/metabolismo , Receptor Tipo 2 de Angiotensina/genética , Receptor Tipo 2 de Angiotensina/metabolismo , Artéria Renal/metabolismo , Artéria Renal/fisiopatologia , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/metabolismo
11.
Am J Physiol Endocrinol Metab ; 310(10): E797-810, 2016 05 15.
Artigo em Inglês | MEDLINE | ID: mdl-26860983

RESUMO

The placental metabolism can adapt to the environment throughout pregnancy to both the demands of the fetus and the signals from the mother. Such adaption processes include epigenetic mechanisms, which alter gene expression and may influence the offspring's health. These mechanisms are linked to the diversity of prenatal environmental exposures, including maternal under- or overnutrition or gestational diabetes. The peroxisome proliferator-activated receptors (PPARs) are nuclear receptors that contribute to the developmental plasticity of the placenta by regulating lipid and glucose metabolism pathways, including lipogenesis, steroidogenesis, glucose transporters, and placental signaling pathways, thus representing a link between energy metabolism and reproduction. Among the PPAR isoforms, PPARγ appears to be the main modulator of mammalian placentation. Certain fatty acids and lipid-derived moieties are the natural activating PPAR ligands. By controlling the amounts of maternal nutrients that go across to the fetus, the PPARs play an important regulatory role in placenta metabolism, thereby adapting to the maternal nutritional status. As demonstrated in animal studies, maternal nutrition during gestation can exert long-term influences on the PPAR methylation pattern in offspring organs. This review underlines the current state of knowledge on the relationship between environmental factors and the epigenetic regulation of the PPARs in placenta metabolism and offspring development.


Assuntos
Diabetes Gestacional/genética , Epigênese Genética , Desenvolvimento Fetal/genética , Desnutrição/genética , Hipernutrição/genética , Receptores Ativados por Proliferador de Peroxissomo/genética , Placenta/metabolismo , Complicações na Gravidez/genética , Diabetes Gestacional/metabolismo , Metabolismo Energético/genética , Ácidos Graxos/metabolismo , Feminino , Feto/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Glucose/metabolismo , Proteínas Facilitadoras de Transporte de Glucose/metabolismo , Humanos , Metabolismo dos Lipídeos/genética , Desnutrição/metabolismo , Hipernutrição/metabolismo , PPAR gama/genética , PPAR gama/metabolismo , Receptores Ativados por Proliferador de Peroxissomo/metabolismo , Placentação , Gravidez , Complicações na Gravidez/metabolismo , Transdução de Sinais
12.
Cell Physiol Biochem ; 39(3): 919-38, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27513464

RESUMO

The concept of developmental origins of diseases has gained a huge interest in recent years and is a constantly emerging scientific field. First observations hereof originated from epidemiological studies, linking impaired birth outcomes to adult chronic, noncommunicable disease. By now there is a considerable amount of both epidemiological and experimental evidence highlighting the impact of early life events on later life disease susceptibility. Albeit far from being completely understood, more recent studies managed to elucidate underlying mechanisms, with epigenetics having become almost synonymous with developmental programming. The aim of this review was to give a comprehensive overview of various aspects and mechanisms of developmental origins of diseases. Starting from initial research foci mainly centered on a nutritionally impaired intrauterine environment, more recent findings such as postnatal nutrition, preterm birth, paternal programming and putative interventional approaches are summarized. The review outlines general underlying mechanisms and particularly discusses mechanistic explanations for sexual dimorphism in developmental programming. Furthermore, novel hypotheses are presented emphasizing a non-mendelian impact of parental genes on the offspring's phenotype.


Assuntos
Epigênese Genética , Regulação da Expressão Gênica no Desenvolvimento , Nascimento Prematuro/genética , Efeitos Tardios da Exposição Pré-Natal/genética , Fenômenos Fisiológicos da Nutrição Pré-Natal/genética , Adulto , Suscetibilidade a Doenças , Feminino , Humanos , Recém-Nascido , Desnutrição/genética , Desnutrição/metabolismo , Desnutrição/fisiopatologia , Hipernutrição/genética , Hipernutrição/metabolismo , Hipernutrição/fisiopatologia , Herança Paterna , Fenótipo , Gravidez , Nascimento Prematuro/metabolismo , Nascimento Prematuro/fisiopatologia , Efeitos Tardios da Exposição Pré-Natal/metabolismo , Efeitos Tardios da Exposição Pré-Natal/fisiopatologia , Caracteres Sexuais
13.
Dig Dis ; 34(4): 391-5, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27170394

RESUMO

In the past 2 decades, advances in genetics have improved our understanding of liver disease and physiology. Firstly, developments in genomic technologies drove the identification of genes responsible for monogenic (Mendelian) liver diseases. Over the last decade, genome-wide association studies allowed for the dissection of the genetic susceptibility to complex liver diseases such as fatty liver disease and drug-induced liver injury, in which environmental co-factors play critical roles. The findings have allowed the identification and elaboration of pathophysiological processes, have indicated the need for reclassification of liver diseases and risk factors and have already pointed to new disease treatments. This is illustrated by the interaction of alcohol, overnutrition and the PNPLA3 gene, which represents an 'infernal triangle' for the liver. In the future, genetics will allow further stratification of liver diseases and contribute to personalized (precision) medicine, offering novel opportunities for translational research and clinical care of our patients.


Assuntos
Hepatopatias/genética , Hepatopatias/terapia , Medicina de Precisão/tendências , Alcoolismo/complicações , Alcoolismo/genética , Doença Hepática Induzida por Substâncias e Drogas/genética , Doença Hepática Induzida por Substâncias e Drogas/terapia , Predisposição Genética para Doença , Estudo de Associação Genômica Ampla , Genômica , Humanos , Lipase/metabolismo , Hepatopatias/fisiopatologia , Proteínas de Membrana/metabolismo , Hipernutrição/complicações , Hipernutrição/genética , Fatores de Risco
14.
Lipids Health Dis ; 14: 96, 2015 Aug 26.
Artigo em Inglês | MEDLINE | ID: mdl-26302954

RESUMO

BACKGROUND: Increasing evidence suggests that overnutrition during the early postnatal period, a critical window of development, increases the risk of adult-onset obesity and insulin resistance. In this study, we investigated the impact of overnutrition during the suckling period on body weight, serum biochemistry and serum fatty acid metabolomics in male rats. METHODS: Rats raised in small litters (SL, 3 pups/dam) and normal litters (NL, 10 pups/dam) were used to model early postnatal overnutrition and control, respectively. Serum glucose, triglyceride, high-density lipoprotein-cholesterol, free fatty acid, insulin and leptin concentrations were assayed using standard biochemical techniques. Serum fatty acids were identified and quantified using a gas chromatography-mass spectrometry-based metabolomic approach. mRNA and protein levels of key components of the insulin receptor signaling pathway were measured in epididymal fat and gastrocnemius muscle by quantitative PCR and western blotting. RESULTS: SL rats were 37.3 % and 15.1 % heavier than NL rats at weaning and 16-weeks-old, respectively. They had increased visceral fat mass, adult-onset insulin resistance and glucose intolerance as well as elevated serum levels of free fatty acids and triglycerides. All detectable fatty acids were elevated in the serum of SL pups at weaning compared to NL controls, and significant increases in the levels of four fatty acids (palmitic acid, palmitoleic acid, oleic acid and arachidonic acid) persisted into adulthood. Moreover, a significantly positive correlation was identified between an insulin resistance index (HOMA-IR) and concentrations of myristic, palmitic, palmitoleic and oleic acid in serum at postnatal 16 weeks. Early postnatal overnutrition also resulted in a significant downregulation of insulin receptor substrate-1 (Irs-1), protein kinase B (Akt2) and glucose transporter 4 (Glut4) at the protein level in epididymal fat of SL rats at 16 weeks, accompanied by decreased mRNA levels for Irs-1 and Glut4. In gastrocnemius muscle, Akt2 and Glut4 mRNA and Glut4 protein levels were significantly decreased in SL rats. CONCLUSIONS: This study demonstrates that early postnatal overnutrition can have long-lasting effects on body weight and serum fatty acid profiles and can lead to impaired insulin signaling pathway in visceral white adipose tissue and skeletal muscle, which may play a major role in IR.


Assuntos
Ácidos Graxos não Esterificados/sangue , Resistência à Insulina , Obesidade/genética , Hipernutrição/genética , RNA Mensageiro/sangue , Tecido Adiposo Branco/metabolismo , Animais , Glicemia/metabolismo , Regulação da Expressão Gênica , Transportador de Glucose Tipo 4/sangue , Transportador de Glucose Tipo 4/genética , Humanos , Insulina/sangue , Insulina/genética , Proteínas Substratos do Receptor de Insulina/sangue , Proteínas Substratos do Receptor de Insulina/genética , Leptina/sangue , Leptina/genética , Lipoproteínas HDL/sangue , Tamanho da Ninhada de Vivíparos , Masculino , Obesidade/sangue , Hipernutrição/sangue , Proteínas Proto-Oncogênicas c-akt/sangue , Proteínas Proto-Oncogênicas c-akt/genética , RNA Mensageiro/genética , Ratos , Ratos Sprague-Dawley , Transdução de Sinais , Triglicerídeos/sangue
15.
Genes (Basel) ; 15(6)2024 May 28.
Artigo em Inglês | MEDLINE | ID: mdl-38927640

RESUMO

This study investigated the transcriptomic responses of subcutaneous adipose tissue (SAT) and liver in newborn Hanwoo calves subjected to maternal overnutrition during mid- to late gestation. Eight Hanwoo cows were randomly assigned to control and treatment groups. The treatment group received a diet of 4.5 kg of concentrate and 6.5 kg of rice straw daily, resulting in intake levels of 8.42 kg DMI, 5.69 kg TDN, and 0.93 kg CP-higher than the control group (6.07 kg DMI, 4.07 kg TDN, and 0.65 kg CP), with respective NEm values of 9.56 Mcal and 6.68 Mcal. Following birth, newly born calves were euthanized humanely as per ethical guidelines, and SAT and liver samples from newborn calves were collected for RNA extraction and analysis. RNA sequencing identified 192 genes that were differentially expressed in the SAT (17 downregulated and 175 upregulated); notably, HSPA6 emerged as the most significantly upregulated gene in the SAT and as the singular upregulated gene in the liver (adj-p value < 0.05). Additionally, differential gene expression analysis highlighted extensive changes across genes associated with adipogenesis, fibrogenesis, and stress response. The functional enrichment pathway and protein-protein interaction (PPI) unraveled the intricate networks and biological processes impacted by overnutrition, including extracellular matrix organization, cell surface receptor signaling, and the PI3K-Akt signaling pathway. These findings underscore maternal overnutrition's substantial influence on developmental pathways, suggesting profound cellular modifications with potential lasting effects on health and productivity. Despite the robust insights that are provided, the study's limitations (sample size) underscore the necessity for further research.


Assuntos
Animais Recém-Nascidos , Fígado , Hipernutrição , Gordura Subcutânea , Transcriptoma , Animais , Feminino , Gravidez , Fígado/metabolismo , Hipernutrição/genética , Bovinos , Gordura Subcutânea/metabolismo , Perfilação da Expressão Gênica/métodos
16.
Sci Rep ; 14(1): 23298, 2024 10 07.
Artigo em Inglês | MEDLINE | ID: mdl-39375502

RESUMO

Maternal nutrition significantly influences fetal development and postnatal outcomes. This study investigates the impact of maternal overfeeding during mid to late pregnancy on gene expression in the round and sirloin muscles of Hanwoo neonatal calves. Eight cows were assigned to either a control group receiving standard nutrition (100%) or a treated group receiving overnutrition (150%). After birth, tissue samples from the round and sirloin muscles of neonatal calves were collected and subjected to RNA sequencing to assess differentially expressed genes (DEGs). RNA sequencing identified 43 DEGs in round muscle and 15 in sirloin muscle, involving genes related to myogenesis, adipogenesis, and energy regulation. Key genes, including PPARGC1A, THBS1, CD44, JUND, CNN1, ENAH, and RUNX1, were predominantly downregulated. Gene ontology (GO) enrichment analyses revealed terms associated with muscle development, such as "biological regulation," "cellular process," and "response to stimulus." Protein-protein interaction networks highlighted complex interactions among DEGs. Random Forest analysis identified ARC, SLC1A5, and GNPTAB as influential genes for distinguishing between control and treated groups. Overall, maternal overnutrition during mid-to-late pregnancy results in the downregulation of genes involved in muscle development and energy metabolism in neonatal Hanwoo calves. These findings provide insights into the molecular effects of maternal nutrition on muscle development.


Assuntos
Animais Recém-Nascidos , Músculo Esquelético , Animais , Bovinos , Gravidez , Feminino , Músculo Esquelético/metabolismo , Desenvolvimento Muscular/genética , Hipernutrição/genética , Hipernutrição/metabolismo , Mapas de Interação de Proteínas , Ontologia Genética , Perfilação da Expressão Gênica , Fenômenos Fisiológicos da Nutrição Materna , Transcriptoma
17.
Br J Nutr ; 107(7): 964-78, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21861941

RESUMO

In addition to being a risk factor for adverse outcomes of pregnancy, maternal obesity may play a role in determining the long-term disease patterns observed in the resulting offspring, with metabolic and dietary factors directly programming fetal development. The present study evaluated the potential for feeding rats an obesogenic cafeteria diet (O) pre-pregnancy, during pregnancy, during lactation and for the offspring post-weaning, to programme glucose tolerance. Early-life exposure to an O diet had no significant effect on offspring food intake. Early-life programming associated with O feeding to induce maternal obesity was associated with reduced adiposity in offspring weaned onto low-fat chow. Adult offspring exposed to an O diet in early life and weaned on a chow diet had low fasting glucose and insulin concentrations and appeared to be more sensitive to insulin during an intraperitoneal glucose tolerance test. When weaned on an O diet, male offspring were more prone to glucose intolerance than females. On the basis of the area under the glucose curve, maternal O feeding at any point from pre-mating to lactation was associated with impaired glucose tolerance. The mechanism for this was not identified, although increased hepatic expression of Akt2 may have indicated disturbance of insulin signalling pathways. The observations in the present study confirm that maternal overnutrition and obesity during pregnancy are risk factors for metabolic disturbance in the resulting offspring. Although the effects on glucose homeostasis were independent of offspring adiposity, the programming of a glucose-intolerant phenotype was only observed when offspring were weaned on a diet that induced greater fat deposition.


Assuntos
Dieta/efeitos adversos , Intolerância à Glucose/etiologia , Fenômenos Fisiológicos da Nutrição Animal , Animais , Modelos Animais de Doenças , Feminino , Intolerância à Glucose/genética , Intolerância à Glucose/metabolismo , Humanos , Proteínas Substratos do Receptor de Insulina/genética , Masculino , Fenômenos Fisiológicos da Nutrição Materna , Obesidade/complicações , Obesidade/genética , Obesidade/metabolismo , Hipernutrição/complicações , Hipernutrição/genética , Hipernutrição/metabolismo , Gravidez , Efeitos Tardios da Exposição Pré-Natal , Proteínas Proto-Oncogênicas c-akt/genética , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Ratos , Ratos Wistar , Fatores de Risco , Desmame
18.
J Nutr Biochem ; 110: 109119, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-35933021

RESUMO

Overnutrition-induced obesity and metabolic dysregulation are considered major risk factors contributing to breast cancer. The origin of both obesity and breast cancer can retrospect to early development in human lifespan. Genistein (GE), a natural isoflavone enriched in soybean products, has been proposed to associate with a lower risk of breast cancer and various metabolic disorders. Our study aimed to determine the effects of maternal exposure to soybean dietary GE on prevention of overnutrition-induced breast cancer later in life and explore potential mechanisms in different mouse models. Our results showed that maternal dietary GE treatment improved offspring metabolic functions by significantly attenuating high-fat diet-induced body fat accumulation, lipid panel abnormalities and glucose intolerance in mice offspring. Importantly, maternal dietary GE exposure effectively delayed high-fat diet-simulated mammary tumor development in female offspring. Mechanistically, we found that maternal dietary GE may exert its chemopreventive effects through affecting essential regulatory gene expression in control of metabolism, inflammation and tumor development via, at least in part, regulation of offspring gut microbiome, bacterial metabolites and epigenetic profiles. Altogether, our findings indicate that maternal GE consumption is an effective intervention approach leading to early-life prevention of obesity-related metabolic disorders and breast cancer later in life through dynamically influencing the interplay between early-life gut microbiota, key microbial metabolite profiles and offspring epigenome.


Assuntos
Microbioma Gastrointestinal , Doenças Metabólicas , Neoplasias , Hipernutrição , Humanos , Camundongos , Feminino , Animais , Glycine max , Epigênese Genética , Obesidade/metabolismo , Dieta Hiperlipídica/efeitos adversos , Hipernutrição/genética , Genisteína/farmacologia , Doenças Metabólicas/genética , Neoplasias/genética
19.
Artigo em Inglês | MEDLINE | ID: mdl-33865538

RESUMO

Using a rabbit model, we investigated whether maternal intake of a high-fat and high-carbohydrate diet (HFCD) before and during pregnancy induces an increase in micronuclei frequency and oxidative stress in offspring during adulthood. Female rabbits received a standard diet (SD) or HFCD for two months before mating and during gestation. The offspring from both groups were nursed by foster mothers fed SD until postnatal day 35. After weaning, all the animals received SD until postnatal day 440. At postnatal day 370, the frequency of micronuclei in peripheral blood reticulocytes (MN-RETs) increased in the male offspring from HFCD-fed mothers compared with the male offspring from SD-fed mothers. Additionally, fasting serum glucose increased in the offspring from HFCD-fed mothers compared with the offspring from SD-fed mothers. At postnatal day 440, the offspring rabbits were challenged with HFCD or continued with SD for 30 days. There was an increase in MN-RET frequency in the male rabbits from HFCD-fed mothers, independent of the type of challenging diet consumed during adulthood. The challenge induced changes in serum cholesterol, LDL and HDL that were influenced by the maternal diet and offspring sex. We measured malondialdehyde in the liver of rabbits as an oxidative stress marker after diet challenge. Oxidative stress in the liver only increased in the female offspring from HFCD-fed mothers who were also challenged with this same diet. The data indicate that maternal overnutrition before and during pregnancy is able to promote different effects depending on the sex of the animals, with chromosomal instability in male offspring and oxidative stress and hypercholesterolemia in female offspring. Our data might be important in the understanding of chronic diseases that develop in adulthood due to in utero exposure to maternal diet.


Assuntos
Dano ao DNA , Hipernutrição/genética , Efeitos Tardios da Exposição Pré-Natal/genética , Animais , Dieta Hiperlipídica/efeitos adversos , Modelos Animais de Doenças , Feminino , Masculino , Fenômenos Fisiológicos da Nutrição Materna , Hipernutrição/complicações , Hipernutrição/patologia , Estresse Oxidativo/fisiologia , Gravidez , Complicações na Gravidez/genética , Complicações na Gravidez/patologia , Efeitos Tardios da Exposição Pré-Natal/patologia , Coelhos , Caracteres Sexuais
20.
Genes (Basel) ; 12(11)2021 10 20.
Artigo em Inglês | MEDLINE | ID: mdl-34828259

RESUMO

Maternal obesity is a rapidly evolving universal epidemic leading to acute and long-term medical and obstetric health issues, including increased maternal risks of gestational diabetes, hypertension and pre-eclampsia, and the future risks for offspring's predisposition to metabolic diseases. Epigenetic modification, in particular DNA methylation, represents a mechanism whereby environmental effects impact on the phenotypic expression of human disease. Maternal obesity or overnutrition contributes to the alterations in DNA methylation during early life which, through fetal programming, can predispose the offspring to many metabolic and chronic diseases, such as non-alcoholic fatty liver disease, obesity, diabetes, and chronic kidney disease. This review aims to summarize findings from human and animal studies, which support the role of maternal obesity in fetal programing and the potential benefit of altering DNA methylation to limit maternal obesity related disease in the offspring.


Assuntos
Doença Crônica , Metilação de DNA/fisiologia , Doenças Metabólicas/genética , Obesidade Materna/genética , Efeitos Tardios da Exposição Pré-Natal , Animais , Suscetibilidade a Doenças , Feminino , Desenvolvimento Fetal/genética , Humanos , Recém-Nascido , Masculino , Obesidade Materna/complicações , Obesidade Materna/metabolismo , Hipernutrição/complicações , Hipernutrição/genética , Hipernutrição/metabolismo , Gravidez , Complicações na Gravidez/genética , Complicações na Gravidez/metabolismo , Efeitos Tardios da Exposição Pré-Natal/genética , Efeitos Tardios da Exposição Pré-Natal/metabolismo , Efeitos Tardios da Exposição Pré-Natal/patologia , Fatores de Risco
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA