Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Traffic ; 25(1): e12920, 2024 01.
Artigo em Inglês | MEDLINE | ID: mdl-37886910

RESUMO

Wilson disease (WD) is caused by mutations in the ATP7B gene that encodes a copper (Cu) transporting ATPase whose trafficking from the Golgi to endo-lysosomal compartments drives sequestration of excess Cu and its further excretion from hepatocytes into the bile. Loss of ATP7B function leads to toxic Cu overload in the liver and subsequently in the brain, causing fatal hepatic and neurological abnormalities. The limitations of existing WD therapies call for the development of new therapeutic approaches, which require an amenable animal model system for screening and validation of drugs and molecular targets. To achieve this objective, we generated a mutant Caenorhabditis elegans strain with a substitution of a conserved histidine (H828Q) in the ATP7B ortholog cua-1 corresponding to the most common ATP7B variant (H1069Q) that causes WD. cua-1 mutant animals exhibited very poor resistance to Cu compared to the wild-type strain. This manifested in a strong delay in larval development, a shorter lifespan, impaired motility, oxidative stress pathway activation, and mitochondrial damage. In addition, morphological analysis revealed several neuronal abnormalities in cua-1 mutant animals exposed to Cu. Further investigation suggested that mutant CUA-1 is retained and degraded in the endoplasmic reticulum, similarly to human ATP7B-H1069Q. As a consequence, the mutant protein does not allow animals to counteract Cu toxicity. Notably, pharmacological correctors of ATP7B-H1069Q reduced Cu toxicity in cua-1 mutants indicating that similar pathogenic molecular pathways might be activated by the H/Q substitution and, therefore, targeted for rescue of ATP7B/CUA-1 function. Taken together, our findings suggest that the newly generated cua-1 mutant strain represents an excellent model for Cu toxicity studies in WD.


Assuntos
Degeneração Hepatolenticular , Animais , Humanos , Degeneração Hepatolenticular/genética , Degeneração Hepatolenticular/tratamento farmacológico , Degeneração Hepatolenticular/metabolismo , Cobre/toxicidade , Cobre/metabolismo , Caenorhabditis elegans/genética , Caenorhabditis elegans/metabolismo , ATPases Transportadoras de Cobre/genética , ATPases Transportadoras de Cobre/metabolismo , Hepatócitos/metabolismo
2.
Proc Natl Acad Sci U S A ; 115(32): 8161-8166, 2018 08 07.
Artigo em Inglês | MEDLINE | ID: mdl-30038027

RESUMO

Copper is an essential cofactor of cytochrome c oxidase (CcO), the terminal enzyme of the mitochondrial respiratory chain. Inherited loss-of-function mutations in several genes encoding proteins required for copper delivery to CcO result in diminished CcO activity and severe pathologic conditions in affected infants. Copper supplementation restores CcO function in patient cells with mutations in two of these genes, COA6 and SCO2, suggesting a potential therapeutic approach. However, direct copper supplementation has not been therapeutically effective in human patients, underscoring the need to identify highly efficient copper transporting pharmacological agents. By using a candidate-based approach, we identified an investigational anticancer drug, elesclomol (ES), that rescues respiratory defects of COA6-deficient yeast cells by increasing mitochondrial copper content and restoring CcO activity. ES also rescues respiratory defects in other yeast mutants of copper metabolism, suggesting a broader applicability. Low nanomolar concentrations of ES reinstate copper-containing subunits of CcO in a zebrafish model of copper deficiency and in a series of copper-deficient mammalian cells, including those derived from a patient with SCO2 mutations. These findings reveal that ES can restore intracellular copper homeostasis by mimicking the function of missing transporters and chaperones of copper, and may have potential in treating human disorders of copper metabolism.


Assuntos
Antineoplásicos/farmacologia , Cobre/deficiência , Drogas em Investigação/farmacologia , Complexo IV da Cadeia de Transporte de Elétrons/metabolismo , Hidrazinas/farmacologia , Mitocôndrias/efeitos dos fármacos , Animais , Antineoplásicos/uso terapêutico , Transporte Biológico/genética , Proteínas de Transporte/genética , Linhagem Celular , Coenzimas/deficiência , Cobre/uso terapêutico , Transportador de Cobre 1 , Suplementos Nutricionais , Modelos Animais de Doenças , Reposicionamento de Medicamentos , Drogas em Investigação/uso terapêutico , Fibroblastos , Humanos , Hidrazinas/uso terapêutico , Proteínas de Membrana Transportadoras/genética , Erros Inatos do Metabolismo/tratamento farmacológico , Erros Inatos do Metabolismo/genética , Erros Inatos do Metabolismo/metabolismo , Mitocôndrias/metabolismo , Proteínas Mitocondriais/genética , Chaperonas Moleculares , Mutagênese Sítio-Dirigida , Mutação , Ratos , Saccharomyces cerevisiae , Peixe-Zebra , Proteínas de Peixe-Zebra/genética
3.
Gastroenterology ; 154(1): 168-180.e5, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-28958857

RESUMO

BACKGROUND & AIMS: Wilson disease is a disorder of copper (Cu) misbalance caused by mutations in ATP7B. ATP7B is highly expressed in the liver-the major site of Cu accumulation in patients with Wilson disease. The intestine also expresses ATP7B, but little is known about the contribution of intestinal ATP7B to normal intestinal copper homeostasis or to Wilson disease manifestations. We characterized the role of ATP7B in mouse intestinal organoids and tissues. METHODS: We collected intestinal tissues from ATP7B-knockout (Atp7b-/-) and control mice, and established 3-dimensional enteroids. Immunohistochemistry and x-ray fluorescence were used to characterize the distribution of ATP7B and Cu in tissues. Electron microscopy, histologic analyses, and immunoblotting were used to determine the effects of ATP7B loss. Enteroids derived from control and ATP7B-knockout mice were incubated with excess Cu or with Cu-chelating reagents; effects on cell fat content and ATP7B levels and localization were determined by fluorescent confocal microscopy. RESULTS: ATP7B maintains a Cu gradient along the duodenal crypt-villus axis and buffers Cu levels in the cytosol of enterocytes. These functions are mediated by rapid Cu-dependent enlargement of ATP7B-containing vesicles and increased levels of ATP7B. Intestines of Atp7b-/- mice had reduced Cu storage pools in intestine, Cu depletion, accumulation of triglyceride-filled vesicles in enterocytes, mislocalization of apolipoprotein B, and loss of chylomicrons. In primary 3-dimensional enteroids, administration of excess Cu or Cu chelators impaired assembly of chylomicrons. CONCLUSIONS: ATP7B regulates vesicular storage of Cu in mouse intestine. ATP7B buffers Cu levels in enterocytes to maintain a range necessary for formation of chylomicrons. Misbalance of Cu and lipid in the intestine could account for gastrointestinal manifestations of Wilson disease.


Assuntos
ATPases Transportadoras de Cobre/metabolismo , Degeneração Hepatolenticular/etiologia , Degeneração Hepatolenticular/metabolismo , Intestinos/enzimologia , Animais , Modelos Animais de Doenças , Feminino , Degeneração Hepatolenticular/patologia , Intestinos/patologia , Masculino , Camundongos , Camundongos Knockout
4.
Sci Rep ; 7(1): 12001, 2017 09 20.
Artigo em Inglês | MEDLINE | ID: mdl-28931909

RESUMO

Copper (Cu) is an essential cofactor for various enzymatic activities including mitochondrial electron transport, iron mobilization, and peptide hormone maturation. Consequently, Cu dysregulation is associated with fatal neonatal disease, liver and cardiac dysfunction, and anemia. While the Cu transporter ATP7A plays a major role in both intestinal Cu mobilization to the periphery and prevention of Cu over-accumulation, it is unclear how regulation of ATP7A contributes to Cu homeostasis in response to systemic Cu fluctuation. Here we show, using Cu-deficient mouse models, that steady-state levels of ATP7A are lower in peripheral tissues (including the heart, spleen, and liver) under Cu deficiency and that subcutaneous administration of Cu to these animals restore normal ATP7A levels in these tissues. Strikingly, ATP7A in the intestine is regulated in the opposite manner - low systemic Cu increases ATP7A while subcutaneous Cu administration decreases ATP7A suggesting that intestine-specific non-autonomous regulation of ATP7A abundance may serve as a key homeostatic control for Cu export into the circulation. Our results support a systemic model for how a single transporter can be inversely regulated in a tissue-specific manner to maintain organismal Cu homeostasis.


Assuntos
ATPases Transportadoras de Cobre/metabolismo , Cobre/metabolismo , Homeostase , Especificidade de Órgãos , Animais , Proteínas de Transporte de Cátions/genética , Proteínas de Transporte de Cátions/metabolismo , Células Cultivadas , Cobre/deficiência , Cobre/farmacologia , Transportador de Cobre 1 , ATPases Transportadoras de Cobre/genética , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/metabolismo , Humanos , Mucosa Intestinal/citologia , Mucosa Intestinal/metabolismo , Fígado/citologia , Fígado/metabolismo , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fenantrolinas/farmacologia , Ratos
5.
J Biol Chem ; 292(1): 1-14, 2017 Jan 06.
Artigo em Inglês | MEDLINE | ID: mdl-27881675

RESUMO

Copper plays key catalytic and regulatory roles in biochemical processes essential for normal growth, development, and health. Defects in copper metabolism cause Menkes and Wilson's disease, myeloneuropathy, and cardiovascular disease and are associated with other pathophysiological states. Consequently, it is critical to understand the mechanisms by which organisms control the acquisition, distribution, and utilization of copper. The intestinal enterocyte is a key regulatory point for copper absorption into the body; however, the mechanisms by which intestinal cells transport copper to maintain organismal copper homeostasis are poorly understood. Here, we identify a mechanism by which organismal copper homeostasis is maintained by intestinal copper exporter trafficking that is coordinated with extraintestinal copper levels in Caenorhabditis elegans Specifically, we show that CUA-1, the C. elegans homolog of ATP7A/B, localizes to lysosome-like organelles (gut granules) in the intestine under copper overload conditions for copper detoxification, whereas copper deficiency results in a redistribution of CUA-1 to basolateral membranes for copper efflux to peripheral tissues. Worms defective in gut granule biogenesis exhibit defects in copper sequestration and increased susceptibility to toxic copper levels. Interestingly, however, a splice isoform CUA-1.2 that lacks a portion of the N-terminal domain is targeted constitutively to the basolateral membrane irrespective of dietary copper concentration. Our studies establish that CUA-1 is a key intestinal copper exporter and that its trafficking is regulated to maintain systemic copper homeostasis. C. elegans could therefore be exploited as a whole-animal model system to study regulation of intra- and intercellular copper trafficking pathways.


Assuntos
Adenosina Trifosfatases/metabolismo , Proteínas de Caenorhabditis elegans/metabolismo , Caenorhabditis elegans/metabolismo , Proteínas de Transporte de Cátions/metabolismo , Membrana Celular/metabolismo , Cobre/metabolismo , Homeostase , Mucosa Intestinal/metabolismo , Animais , Caenorhabditis elegans/crescimento & desenvolvimento , Intestinos/crescimento & desenvolvimento , Transporte Proteico
6.
Mol Cell Biol ; 32(7): 1284-95, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22290441

RESUMO

Copper (Cu) is essential for development and proliferation, yet the cellular requirements for Cu in these processes are not well defined. We report that Cu plays an unanticipated role in the mitogen-activated protein (MAP) kinase pathway. Ablation of the Ctr1 high-affinity Cu transporter in flies and mouse cells, mutation of Ctr1, and Cu chelators all reduce the ability of the MAP kinase kinase Mek1 to phosphorylate the MAP kinase Erk. Moreover, mice bearing a cardiac-tissue-specific knockout of Ctr1 are deficient in Erk phosphorylation in cardiac tissue. in vitro investigations reveal that recombinant Mek1 binds two Cu atoms with high affinity and that Cu enhances Mek1 phosphorylation of Erk in a dose-dependent fashion. Coimmunoprecipitation experiments suggest that Cu is important for promoting the Mek1-Erk physical interaction that precedes the phosphorylation of Erk by Mek1. These results demonstrate a role for Ctr1 and Cu in activating a pathway well known to play a key role in normal physiology and in cancer.


Assuntos
Proteínas de Transporte de Cátions/metabolismo , Cobre/metabolismo , Sistema de Sinalização das MAP Quinases , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Proteínas ras/metabolismo , Animais , Proteínas de Transporte de Cátions/genética , Linhagem Celular , Transportador de Cobre 1 , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/genética , Drosophila melanogaster/metabolismo , Ativação Enzimática , Deleção de Genes , MAP Quinase Quinase 1/metabolismo , Camundongos , Fosforilação , Ligação Proteica
7.
J Biol Chem ; 285(42): 32385-92, 2010 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-20699218

RESUMO

Copper is an essential trace element that functions in a diverse array of biochemical processes that include mitochondrial respiration, neurotransmitter biogenesis, connective tissue maturation, and reactive oxygen chemistry. The Ctr1 protein is a high-affinity Cu(+) importer that is structurally and functionally conserved in yeast, plants, fruit flies, and humans and that, in all of these organisms, is localized to the plasma membrane and intracellular vesicles. Although intestinal epithelial cell-specific deletion of Ctr1 in mice demonstrated a critical role for Ctr1 in dietary copper absorption, some controversy exists over the localization of Ctr1 in intestinal epithelial cells in vivo. In this work, we assess the localization of Ctr1 in intestinal epithelial cells through two independent mechanisms. Using immunohistochemistry, we demonstrate that Ctr1 localizes to the apical membrane in intestinal epithelial cells of the mouse, rat, and pig. Moreover, biotinylation of intestinal luminal proteins from mice fed a control or a copper-deficient diet showed elevated levels of both total and apical membrane Ctr1 protein in response to transient dietary copper limitation. Experiments in cultured HEK293T cells demonstrated that alterations in the levels of the glycosylated form of Ctr1 in response to copper availability were a time-dependent, copper-specific posttranslational response. Taken together, these results demonstrate apical localization of Ctr1 in intestinal epithelia across three mammalian species and suggest that increased Ctr1 apical localization in response to dietary copper limitation may represent an adaptive response to homeostatically modulate Ctr1 availability at the site of intestinal copper absorption.


Assuntos
Proteínas de Transporte de Cátions/metabolismo , Cobre/metabolismo , Células Epiteliais/metabolismo , Mucosa Intestinal/citologia , Estabilidade Proteica , Sequência de Aminoácidos , Animais , Proteínas de Transporte de Cátions/genética , Polaridade Celular , Transportador de Cobre 1 , Dieta , Células Epiteliais/citologia , Humanos , Mucosa Intestinal/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Dados de Sequência Molecular , Ratos , Suínos
8.
Cell Metab ; 11(5): 353-63, 2010 May 05.
Artigo em Inglês | MEDLINE | ID: mdl-20444417

RESUMO

Copper (Cu) is an essential cofactor for a variety of metabolic functions, and the regulation of systemic Cu metabolism is critical to human health. Dietary Cu is absorbed through the intestine, stored in the liver, and mobilized into the circulation; however, systemic Cu homeostasis is poorly understood. We generated mice with a cardiac-specific knockout of the Ctr1 Cu transporter (Ctr1(hrt/hrt)), resulting in cardiac Cu deficiency and severe cardiomyopathy. Unexpectedly, Ctr1(hrt/hrt) mice exhibited increased serum Cu levels and a concomitant decrease in hepatic Cu stores. Expression of the ATP7A Cu exporter, thought to function predominantly in intestinal Cu acquisition, was strongly increased in liver and intestine of Ctr1(hrt/hrt) mice. These studies identify ATP7A as a candidate for hepatic Cu mobilization in response to peripheral tissue demand, and illuminate a systemic regulation in which the Cu status of the heart is signaled to organs that take up and store Cu.


Assuntos
Cobre/metabolismo , Miocárdio/metabolismo , Transdução de Sinais , Adenosina Trifosfatases/metabolismo , Animais , Cardiomiopatia Dilatada/etiologia , Proteínas de Transporte de Cátions/deficiência , Proteínas de Transporte de Cátions/genética , Proteínas de Transporte de Cátions/metabolismo , Cobre/deficiência , Transportador de Cobre 1 , ATPases Transportadoras de Cobre , Drosophila/metabolismo , Mucosa Intestinal/metabolismo , Fígado/metabolismo , Camundongos , Camundongos Knockout
9.
J Med Genet ; 44(10): 641-6, 2007 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17483305

RESUMO

Owing to mutations in the copper-transporting P-type ATPase, ATP7A (or MNK), patients with Menkes disease (MD) have an inadequate supply of copper to various copper-dependent enzymes. The ATP7A protein is located in the trans-Golgi network, where it transports copper via secretory compartments to copper-dependent enzymes. Raised copper concentrations result in the trafficking of ATP7A to the plasma membrane, where it functions in copper export. An important model of MD is the Mottled mouse, which possesses mutations in Atp7A. The Mottled mouse displays three distinct phenotypic severities: embryonic lethal, perinatal lethal and a longer-lived viable phenotype. However, the effects of mutations from these phenotypic classes on the ATP7A protein are unknown. In this study, we found that these classes of mutation differentially affect the copper transport and trafficking functions of the ATP7A protein. The embryonic lethal mutation, Atp7a(mo11H) (11H), caused mislocalisation of the protein to the endoplasmic reticulum, impaired glycosylation, and abolished copper delivery to the secretory pathway. In contrast, the perinatal lethal and viable mutations, Atp7a(moMac) (Macular) and Atp7a(moVbr) (Viable brindle) both resulted in a reduction in copper delivery to the secretory pathway and constitutive trafficking of the ATP7A protein to the plasma membrane in the absence of additional copper. In the case of Viable brindle, this hypertrafficking response was dependent on the catalytic phosphorylation site of ATP7A, whereas no such requirement was found for the Macular mutation. These findings provide evidence that the degree of MD severity in mice is associated with both copper transport and trafficking defects in the ATP7A protein.


Assuntos
Adenosina Trifosfatases/genética , Adenosina Trifosfatases/fisiologia , Proteínas de Transporte de Cátions/genética , Proteínas de Transporte de Cátions/fisiologia , Síndrome dos Cabelos Torcidos/genética , Animais , ATPases Transportadoras de Cobre , Retículo Endoplasmático/metabolismo , Complexo de Golgi/metabolismo , Camundongos , Microscopia de Fluorescência , Modelos Biológicos , Modelos Genéticos , Mutação , Fenótipo , Fosforilação , Plasmídeos/metabolismo , Isomerases de Dissulfetos de Proteínas/metabolismo , Transporte Proteico
10.
Cell Metab ; 4(3): 235-44, 2006 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16950140

RESUMO

The trace element copper (Cu) is a cofactor for biochemical functions ranging from energy generation to iron (Fe) acquisition, angiogenesis, and free radical detoxification. While Cu is essential for life, the molecules that mediate dietary Cu uptake have not been identified. Ctr1 is a homotrimeric protein, conserved from yeast to humans, that transports Cu across the plasma membrane with high affinity and specificity. Here we describe the generation of intestinal epithelial cell-specific Ctr1 knockout mice. These mice exhibit striking neonatal defects in Cu accumulation in peripheral tissues, hepatic Fe overload, cardiac hypertrophy, and severe growth and viability defects. Consistent with an intestinal Cu absorption block, the growth and viability defects can be partially rescued by a single postnatal Cu administration, indicative of a critical neonatal metabolic requirement for Cu that is provided by intestinal Ctr1. These studies identify Ctr1 as the major factor driving intestinal Cu absorption in mammals.


Assuntos
Proteínas de Transporte de Cátions/metabolismo , Cobre/deficiência , Absorção Intestinal/genética , Mucosa Intestinal/metabolismo , Ferro/metabolismo , Proteínas de Membrana Transportadoras/metabolismo , Animais , Animais Recém-Nascidos , Cardiomegalia/genética , Cardiomegalia/metabolismo , Cardiomegalia/fisiopatologia , Proteínas de Transporte de Cátions/genética , Membrana Celular/metabolismo , Cobre/farmacologia , Transportador de Cobre 1 , Modelos Animais de Doenças , Nanismo/genética , Nanismo/metabolismo , Nanismo/fisiopatologia , Cardiopatias Congênitas/genética , Cardiopatias Congênitas/metabolismo , Cardiopatias Congênitas/fisiopatologia , Mucosa Intestinal/fisiopatologia , Intestinos/fisiopatologia , Síndromes de Malabsorção/genética , Síndromes de Malabsorção/metabolismo , Síndromes de Malabsorção/fisiopatologia , Proteínas de Membrana Transportadoras/genética , Camundongos , Camundongos Knockout
11.
J Biol Chem ; 277(48): 46736-42, 2002 Nov 29.
Artigo em Inglês | MEDLINE | ID: mdl-12228238

RESUMO

The Menkes protein (MNK; ATP7A) is a copper-transporting P-type ATPase that is defective in the copper deficiency disorder, Menkes disease. MNK is localized in the trans-Golgi network and transports copper to enzymes synthesized within secretory compartments. However, in cells exposed to excessive copper, MNK traffics to the plasma membrane where it functions in copper efflux. A conserved feature of all P-type ATPases is the formation of an acyl-phosphate intermediate, which occurs as part of the catalytic cycle during cation transport. In this study we investigated the effect of mutations within conserved catalytic regions of MNK on intracellular localization and trafficking from the trans-Golgi network (TGN). Our findings suggest that mutations that block formation of the phosphorylated catalytic intermediate also prevent copper-induced relocalization of MNK from the TGN. Furthermore, mutations in the phosphatase domain, which resulted in hyperphosphorylation of MNK, caused constitutive trafficking from the TGN to the plasma membrane. A similar effect on trafficking was observed with a phosphatase mutation in the closely related copper ATPase, ATP7B, affected in Wilson disease. These findings suggest that the copper-induced trafficking of the Menkes and Wilson disease copper ATPases is associated with the phosphorylated intermediate that is formed during the catalysis of these pumps. Our findings describe a novel mechanism for regulating the subcellular location of a transport protein involving the recognition of intermediate conformations during catalysis.


Assuntos
Adenosina Trifosfatases/metabolismo , Proteínas de Transporte de Cátions/metabolismo , Cobre/fisiologia , Proteínas Recombinantes de Fusão , Adenosina Trifosfatases/genética , Animais , Western Blotting , Células CHO , Catálise , Proteínas de Transporte de Cátions/genética , ATPases Transportadoras de Cobre , Cricetinae , Imunofluorescência , Teste de Complementação Genética , Mutagênese , Fosforilação , Transporte Proteico , Frações Subcelulares/enzimologia
12.
J Biol Chem ; 277(46): 44079-84, 2002 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-12221109

RESUMO

Copper is an essential co-factor for several key metabolic processes. This requirement in humans is underscored by Menkes disease, an X-linked copper deficiency disorder caused by mutations in the copper transporting P-type ATPase, MNK. MNK is located in the trans-Golgi network where it transports copper to secreted cuproenzymes. Increases in copper concentration stimulate the trafficking of MNK to the plasma membrane where it effluxes copper. In this study, a Menkes disease mutation, G1019D, located in the large cytoplasmic loop of MNK, was characterized in transfected cultured cells. In copper-limiting conditions the G1019D mutant protein was retained in the endoplasmic reticulum. However, this mislocalization was corrected by the addition of copper to cells via a process that was dependent upon the copper binding sites at the N-terminal region of MNK. Reduced growth temperature and the chemical chaperone, glycerol, were found to correct the mislocalization of the G1019D mutant, suggesting this mutation interferes with protein folding in the secretory pathway. These findings identify G1019D as the first conditional mutation associated with Menkes disease and demonstrate correction of the mislocalized protein by copper supplementation. Our findings provide a molecular framework for understanding how mutations that affect the proper folding of the MNK transporter in Menkes patients may be responsive to parenteral copper therapy.


Assuntos
Adenosina Trifosfatases/genética , Proteínas de Transporte de Cátions/genética , Cobre/farmacologia , Síndrome dos Cabelos Torcidos/enzimologia , Mutação , Proteínas Recombinantes de Fusão , Adenosina Trifosfatases/química , Sítios de Ligação , Proteínas de Transporte de Cátions/química , Linhagem Celular , Membrana Celular/metabolismo , Cobre/metabolismo , ATPases Transportadoras de Cobre , Retículo Endoplasmático/metabolismo , Fibroblastos/metabolismo , Glicosilação , Humanos , Immunoblotting , Síndrome dos Cabelos Torcidos/genética , Microscopia de Fluorescência , Ligação Proteica , Dobramento de Proteína , Temperatura , Transfecção
13.
J Nutr ; 132(8 Suppl): 2419S-2423S, 2002 08.
Artigo em Inglês | MEDLINE | ID: mdl-12163704

RESUMO

In this study we examine the effect of the phytoestrogen genistein on DNA methylation. DNA methylation is thought to inhibit transcription of genes by regulating alterations in chromatin structure. Estrogenic compounds have been reported to regulate DNA methylation in a small number of studies. Additionally, phytoestrogens are believed to affect progression of some human diseases, such as estrogen-dependent cancers, osteoporosis and cardiovascular disease. Specifically, our working hypothesis is that certain soy phytoestrogens, such as genistein, may be involved in preventing the development of certain prostate and mammary cancers by maintaining a protective DNA methylation profile. The objective of the present study is to use mouse differential methylation hybridization (DMH) arrays to test for changes in the methylation status of the cytosine guanine dinucleotide (CpG) islands in the mouse genome by examining how these methylation patterns are affected by genistein. Male mice were fed a casein-based diet (control) or the same diet containing 300 mg genistein/kg according to one of four regimens: control diet for 4 wk, genistein diet for 4 wk, control diet for 2 wk followed by genistein diet for 2 wk and genistein diet for 2 wk followed by control diet for 2 wk. DNA from liver, brain and prostate were then screened with DMH arrays. Clones with methylation differences were sequenced and compared with known sequences. In conclusion, consumption of genistein diet was positively correlated with changes in prostate DNA methylation at CpG islands of specific mouse genes.


Assuntos
Metilação de DNA , Genisteína/farmacologia , Animais , Sequência de Bases , Ilhas de CpG , DNA , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Hibridização de Ácido Nucleico
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA