Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Diabetologia ; 58(3): 615-24, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25608625

RESUMO

AIMS/HYPOTHESIS: Maternal obesity is associated with an increased risk of obesity and impaired glucose homeostasis in offspring. However, it is not known whether a gestational or pre-gestational exposure confers similar risks, and if so, what the underlying mechanisms are. METHODS: We used reciprocal two-cell embryo transfers between mice fed either a control or high-fat diet (HFD) starting at the time of weaning. Gene expression in placenta was assessed by microarray analyses. RESULTS: A pre-gestational exposure to a maternal HFD (HFD/control) impaired fetal and placental growth despite a normal gestational milieu. Expression of imprinted genes and genes regulating vasculogenesis and lipid metabolism was markedly altered in placenta of HFD/control. An exposure to an HFD (control/HFD) only during gestation also resulted in fetal growth restriction and decreased placental weight. Interestingly, only a gestational exposure to an HFD (control/HFD) resulted in obesity and impaired glucose tolerance in adulthood. CONCLUSIONS/INTERPRETATION: An HFD during pregnancy has profound consequences for the offspring later in life. Our data demonstrate that the mechanism underlying this phenomenon is not related to placental dysfunction, intrauterine growth restriction or postnatal weight gain, but rather an inability of the progeny to adapt to the abnormal gestational milieu of an HFD. Thus, the ability to adapt to an adverse intrauterine environment is conferred prior to pregnancy and it is possible that the effects of a maternal HFD may be transmitted to subsequent generations.


Assuntos
Obesidade/complicações , Animais , Animais Recém-Nascidos , Peso Corporal/fisiologia , Dieta Hiperlipídica/efeitos adversos , Feminino , Retardo do Crescimento Fetal/etiologia , Masculino , Camundongos , Placenta/patologia , Gravidez , Complicações na Gravidez , Efeitos Tardios da Exposição Pré-Natal
2.
Arch Toxicol ; 88(8): 1573-88, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24535564

RESUMO

Liver injury is the leading cause of drug-induced toxicity. For the evaluation of a chemical compound to induce toxicity, in this case steatosis or fatty liver, it is imperative to identify markers reflective of mechanisms and processes induced upon exposure, as these will be the earliest changes reflective of disease. Therefore, an in vivo mouse toxicogenomics study was completed to identify common pathways, nuclear receptor (NR) binding sites, and genes regulated by three known human steatosis-inducing compounds, amiodarone (AMD), valproic acid (VPA), and tetracycline (TET). Over 1, 4, and 11 days of treatment, AMD induced changes in clinical chemistry parameters and histopathology consistent with steatosis. Common processes and NR binding sites involved in lipid, retinol, and drug metabolism were found for AMD and VPA, but not for TET, which showed no response. Interestingly, the pattern of enrichment of these common pathways and NR binding sites over time was unique to each compound. Eleven biomarkers of steatosis were identified as dose responsive and time sensitive to toxicity for AMD and VPA. Finally, this in vivo mouse study was compared to an AMD rat in vivo, an AMD mouse primary hepatocyte, and a VPA human primary hepatocyte study to identify concordance for steatosis. We conclude that concordance is found on the process level independent of species, model or dose*time point.


Assuntos
Amiodarona/toxicidade , Doença Hepática Induzida por Substâncias e Drogas/etiologia , Modelos Animais de Doenças , Fígado Gorduroso/complicações , Receptores Citoplasmáticos e Nucleares/metabolismo , Transcriptoma , Ácido Valproico/toxicidade , Amiodarona/farmacocinética , Animais , Sítios de Ligação , Linhagem Celular , Doença Hepática Induzida por Substâncias e Drogas/genética , Doença Hepática Induzida por Substâncias e Drogas/metabolismo , Doença Hepática Induzida por Substâncias e Drogas/patologia , Relação Dose-Resposta a Droga , Fígado Gorduroso/induzido quimicamente , Fígado Gorduroso/genética , Fígado Gorduroso/metabolismo , Hepatócitos/efeitos dos fármacos , Hepatócitos/metabolismo , Humanos , Testes de Função Hepática , Masculino , Camundongos Endogâmicos C57BL , Ratos , Receptores Citoplasmáticos e Nucleares/genética , Especificidade da Espécie , Ácido Valproico/farmacocinética
3.
Genetics ; 172(3): 1711-26, 2006 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-16387879

RESUMO

Although mutation, genetic drift, and natural selection are well established as determinants of genome evolution, the importance (frequency and magnitude) of parameter fluctuations in molecular evolution is less understood. DNA sequence comparisons among closely related species allow specific substitutions to be assigned to lineages on a phylogenetic tree. In this study, we compare patterns of codon usage and protein evolution in 22 genes (>11,000 codons) among Drosophila melanogaster and five relatives within the D. melanogaster subgroup. We assign changes to eight lineages using a maximum-likelihood approach to infer ancestral states. Uncertainty in ancestral reconstructions is taken into account, at least to some extent, by weighting reconstructions by their posterior probabilities. Four of the eight lineages show potentially genomewide departures from equilibrium synonymous codon usage; three are decreasing and one is increasing in major codon usage. Several of these departures are consistent with lineage-specific changes in selection intensity (selection coefficients scaled to effective population size) at silent sites. Intron base composition and rates and patterns of protein evolution are also heterogeneous among these lineages. The magnitude of forces governing silent, intron, and protein evolution appears to have varied frequently, and in a lineage-specific manner, within the D. melanogaster subgroup.


Assuntos
Drosophila melanogaster/genética , Evolução Molecular , Filogenia , Substituição de Aminoácidos/genética , Animais , Composição de Bases , Códon , Frequência do Gene , Íntrons , Dados de Sequência Molecular , Polimorfismo Genético , Especificidade da Espécie
4.
Toxicol Lett ; 232(2): 403-12, 2015 Jan 22.
Artigo em Inglês | MEDLINE | ID: mdl-25448281

RESUMO

The zebrafish embryo (ZFE) is a promising alternative, non-rodent model in toxicology, which has an advantage over the traditionally used models as it contains complete biological complexity and provides a medium to high-throughput setting. Here, we assess how the ZFE compares to the traditionally used models for liver toxicity testing, i.e., in vivo mouse and rat liver, in vitro mouse and rat hepatocytes, and primary human hepatocytes. For this comparison, we analyzed gene expression changes induced by three model compounds for cholestasis, steatosis, and necrosis. The three compounds, cyclosporine A, amiodarone, and acetaminophen, were chosen because of their relevance to human toxicity and these compounds displayed hepatotoxic-specific changes in the mouse in vivo data. Compound induced expression changes in the ZFE model shared similarity with both in vivo and in vitro. Comparison on single gene level revealed the presence of model specific changes and no clear concordance across models. However, concordance was identified on the pathway level. Specifically, the pathway "regulation of metabolism - bile acids regulation of glucose and lipid metabolism via FXR" was affected across all models and compounds. In conclusion, our study with three hepatotoxic model compounds shows that the ZFE model is at least as comparable to traditional models in identifying hepatotoxic activity and has the potential for use as a pre-screen to determine the hepatotoxic potential of compounds.


Assuntos
Acetaminofen/toxicidade , Amiodarona/toxicidade , Analgésicos não Narcóticos/toxicidade , Antiarrítmicos/toxicidade , Doença Hepática Induzida por Substâncias e Drogas/patologia , Ciclosporina/toxicidade , Embrião não Mamífero/efeitos dos fármacos , Imunossupressores/toxicidade , Transcriptoma/efeitos dos fármacos , Peixe-Zebra/fisiologia , Animais , Expressão Gênica/efeitos dos fármacos , Hepatócitos/efeitos dos fármacos , Humanos , Técnicas In Vitro , Camundongos , Camundongos Endogâmicos C57BL , Ratos
5.
Neuropsychopharmacology ; 39(4): 801-10, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23924601

RESUMO

Maternal obesity during pregnancy increases the risk for offspring obesity, in part through effects on the developing brain. Previous research has shown that perinatal consumption of highly palatable foods by the mother can influence the development of offspring taste preferences and alter gene expression within the central nervous system (CNS) reward system. Opioids stimulate consumption of both fats and carbohydrates, and overconsumption of these energy dense foods increases the risk for obesity. What has remained unclear is whether this risk can be transmitted to the offspring before gestation or if it is wholly the gestational exposure that affects offspring brain development. Utilizing an embryo transfer experimental design, 2-cell embryos were obtained from obese or control dams, and transferred to obese or control gestational carriers. Expression of the mu-opioid receptor (MOR), preproenkephalin (PENK), and the dopamine transporter was evaluated in the hypothalamus and reward circuitry (ventral tegmental area, prefrontal cortex, and nucleus accumbens) in adult and late embryonic brains. Obesity before pregnancy altered expression levels of both MOR and PENK, with males relatively more affected than females. These data are the first to demonstrate that obesity at conception, in addition to during gestation, can program the brain reward system.


Assuntos
Encéfalo/metabolismo , Proteínas da Membrana Plasmática de Transporte de Dopamina/metabolismo , Encefalinas/metabolismo , Obesidade/fisiopatologia , Efeitos Tardios da Exposição Pré-Natal , Precursores de Proteínas/metabolismo , Receptores Opioides mu/metabolismo , Animais , Encéfalo/crescimento & desenvolvimento , Metilação de DNA/genética , Modelos Animais de Doenças , Proteínas da Membrana Plasmática de Transporte de Dopamina/genética , Destinação do Embrião , Encefalinas/genética , Feminino , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Genes sry/genética , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Masculino , Relações Materno-Fetais , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Gravidez , Precursores de Proteínas/genética , Receptores Opioides mu/genética
6.
Toxicol Lett ; 230(1): 48-56, 2014 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-25064622

RESUMO

The zebrafish embryo (ZFE) is a promising non-rodent model in toxicology, and initial studies suggested its applicability in detecting hepatotoxic responses. Here, we hypothesize that the detailed analysis of underlying mechanisms of hepatotoxicity in ZFE contributes to the improved identification of hepatotoxic properties of new compounds and to the reduction of rodents used for screening. ZFEs were exposed to nine reference hepatotoxicants, targeted at induction of cholestasis, steatosis and necrosis, and two non-hepatotoxic controls. Histopathology revealed various specific morphological changes in the ZFE hepatocytes indicative of cell injury. Gene expression profiles of the individual compounds were generated using microarrays. Regulation of single genes and of pathways could be linked to hepatotoxic responses in general, but phenotype-specific responses could not be distinguished. Hepatotoxicity-associated pathways included xenobiotic metabolism and oxidoreduction related pathways. Overall analysis of gene expression identified a limited set of potential biomarkers specific for a common hepatotoxicity response. This set included several cytochrome P450 genes (cyp2k19, cyp4v7, cyp2aa3), genes related to liver development (pklr) and genes important in oxidoreduction processes (zgc:163022, zgc:158614, zgc:101858 and sqrdl). In conclusion, the ZFE model allows for identification of hepatotoxicants, without discrimination into specific phenotypes.


Assuntos
Análise por Conglomerados , Perfilação da Expressão Gênica/métodos , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Fígado/efeitos dos fármacos , Testes de Toxicidade/métodos , Peixe-Zebra/genética , Animais , Redes Reguladoras de Genes , Marcadores Genéticos , Humanos , Fígado/embriologia , Fígado/enzimologia , Fígado/patologia , Modelos Animais , Análise de Sequência com Séries de Oligonucleotídeos , Fenótipo , Medição de Risco , Especificidade da Espécie , Peixe-Zebra/embriologia , Peixe-Zebra/metabolismo
7.
Toxicol Lett ; 221(3): 225-36, 2013 Aug 29.
Artigo em Inglês | MEDLINE | ID: mdl-23830897

RESUMO

In vitro models for hepatotoxicity testing are a necessity for advancement of toxicological research. Assessing the in vitro response requires in vivo validated gene sets reflective of the hepatotoxic phenotype. Cholestasis, the impairment of bile flow, is induced in C57BL/6J mice treated with cyclosporine A (CsA) to identify phenotype reflective gene sets. CsA treatment through oral gavage for 25 days induced cholestasis, as confirmed by histopathology and serum chemistry. Over 1, 4, and 11 days of CsA exposure gradual increases in serum markers were correlated to gene expression. This phenotype-directed analysis identified gene sets specific to the onset and progression of cholestasis, such as PPAR related processes and drug metabolism, by circumventing other effects of CsA, such as immunosuppression, found in dose*time group analysis. In vivo gene sets are enriched in publicly available data sets of CsA-treated HepaRG and primary mouse hepatocytes. However, genes identified within these gene sets did not overlap between in vivo and in vitro. In vitro regulated genes represent the initial response to cholestasis, whereas in vivo genes represent the later adaptive response. We conclude that the applicability of in vitro models for hepatotoxicity testing fully depends on a solid in vivo phenotype anchored analysis.


Assuntos
Colestase/induzido quimicamente , Ciclosporina/toxicidade , Alanina Transaminase/sangue , Animais , Aspartato Aminotransferases/sangue , Ácidos e Sais Biliares/sangue , Bilirrubina/sangue , Colestase/sangue , Colestase/genética , Colesterol/sangue , Expressão Gênica/efeitos dos fármacos , Perfilação da Expressão Gênica/métodos , Células Hep G2 , Histocitoquímica , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Fenótipo , Organismos Livres de Patógenos Específicos , Toxicogenética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA