Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 3 de 3
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Metallomics ; 6(3): 542-53, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24343124

RESUMO

The loss of NPC1 protein function is the predominant cause of Niemann-Pick type C1 disease (NP-C1), a systemic and neurodegenerative disorder characterized by late-endosomal/lysosomal accumulation of cholesterol and other lipids. Limited evidence from post-mortem human tissues, an Npc1(-/-) mouse model, and cell culture studies also suggest failure of metal homeostasis in NP-C1. To investigate these findings, we performed a comprehensive transition metal analysis of cerebrospinal fluid (CSF), plasma and tissue samples from human NP-C1 patients and an Npc1(-/-) mouse model. NPC1 deficiency in the Npc1(-/-) mouse model resulted in a perturbation of transition metal homeostasis in the plasma and key organs (brain, liver, spleen, heart, lungs, and kidneys). Analysis of human patient CSF, plasma and post-mortem brain tissues also indicated disrupted metal homeostasis. There was a disparity in the direction of metal changes between the human and the Npc1(-/-) mouse samples, which may reflect species-specific metal metabolism. Nevertheless, common to both species is brain zinc accumulation. Furthermore, treatment with the glucosylceramide synthase inhibitor miglustat, the only drug shown in a controlled clinical trial to have some efficacy for NP-C1, did not correct the alterations in CSF and plasma transition metal and ceruloplasmin (CP) metabolism in NP-C1 patients. These findings highlight the importance of NPC1 function in metal homeostasis, and indicate that metal-targeting therapy may be of value as a treatment for NP-C.


Assuntos
Metais/metabolismo , Doença de Niemann-Pick Tipo C/metabolismo , Elementos de Transição/metabolismo , 1-Desoxinojirimicina/análogos & derivados , 1-Desoxinojirimicina/uso terapêutico , Animais , Encéfalo/metabolismo , Encéfalo/patologia , Ceruloplasmina/antagonistas & inibidores , Ceruloplasmina/metabolismo , Inibidores Enzimáticos/uso terapêutico , Feminino , Deleção de Genes , Homeostase , Humanos , Peptídeos e Proteínas de Sinalização Intracelular , Masculino , Metais/sangue , Metais/líquido cefalorraquidiano , Camundongos , Camundongos Endogâmicos BALB C , Proteína C1 de Niemann-Pick , Doença de Niemann-Pick Tipo C/sangue , Doença de Niemann-Pick Tipo C/líquido cefalorraquidiano , Doença de Niemann-Pick Tipo C/tratamento farmacológico , Proteínas/genética , Elementos de Transição/sangue , Elementos de Transição/líquido cefalorraquidiano
2.
Hum Mol Genet ; 22(17): 3508-23, 2013 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-23666527

RESUMO

Niemann-Pick disease, type C1 (NPC1), which arises from a mutation in the NPC1 gene, is characterized by abnormal cellular storage and transport of cholesterol and other lipids that leads to hepatic disease and progressive neurological impairment. Oxidative stress has been hypothesized to contribute to the NPC1 disease pathological cascade. To determine whether treatments reducing oxidative stress could alleviate NPC1 disease phenotypes, the in vivo effects of the antioxidant N-acetylcysteine (NAC) on two mouse models for NPC1 disease were studied. NAC was able to partially suppress phenotypes in both antisense-induced (NPC1ASO) and germline (Npc1-/-) knockout genetic mouse models, confirming the presence of an oxidative stress-related mechanism in progression of NPC1 phenotypes and suggesting NAC as a potential molecule for treatment. Gene expression analyses of NAC-treated NPC1ASO mice suggested NAC affects pathways distinct from those initially altered by Npc1 knockdown, data consistent with NAC achieving partial disease phenotype suppression. In a therapeutic trial of short-term NAC administration to NPC1 patients, no significant effects on oxidative stress in these patients were identified other than moderate improvement of the fraction of reduced CoQ10, suggesting limited efficacy of NAC monotherapy. However, the mouse model data suggest that the distinct antioxidant effects of NAC could provide potential treatment of NPC1 disease, possibly in concert with other therapeutic molecules at earlier stages of disease progression. These data also validated the NPC1ASO mouse as an efficient model for candidate NPC1 drug screening, and demonstrated similarities in hepatic phenotypes and genome-wide transcript expression patterns between the NPC1ASO and Npc1-/- models.


Assuntos
Acetilcisteína/farmacologia , Acetilcisteína/uso terapêutico , Doença de Niemann-Pick Tipo C/tratamento farmacológico , Doença de Niemann-Pick Tipo C/genética , Estresse Oxidativo/efeitos dos fármacos , Acetilcisteína/administração & dosagem , Adolescente , Adulto , Animais , Criança , Pré-Escolar , Estudos Cross-Over , Modelos Animais de Doenças , Método Duplo-Cego , Feminino , Expressão Gênica , Humanos , Fígado/metabolismo , Fígado/patologia , Masculino , Camundongos , Camundongos Knockout , Doença de Niemann-Pick Tipo C/metabolismo , Doença de Niemann-Pick Tipo C/fisiopatologia , Estresse Oxidativo/genética , Ubiquinona/análogos & derivados , Ubiquinona/metabolismo , Adulto Jovem
3.
J Biol Chem ; 287(47): 39349-60, 2012 Nov 16.
Artigo em Inglês | MEDLINE | ID: mdl-23035117

RESUMO

Niemann-Pick disease type C (NPC) and Wolman disease are two members of a family of storage disorders caused by mutations of genes encoding lysosomal proteins. Deficiency in function of either the NPC1 or NPC2 protein in NPC disease or lysosomal acid lipase in Wolman disease results in defective cellular cholesterol trafficking. Lysosomal accumulation of cholesterol and enlarged lysosomes are shared phenotypic characteristics of both NPC and Wolman cells. Utilizing a phenotypic screen of an approved drug collection, we found that δ-tocopherol effectively reduced lysosomal cholesterol accumulation, decreased lysosomal volume, increased cholesterol efflux, and alleviated pathological phenotypes in both NPC1 and Wolman fibroblasts. Reduction of these abnormalities may be mediated by a δ-tocopherol-induced intracellular Ca(2+) response and subsequent enhancement of lysosomal exocytosis. Consistent with a general mechanism for reduction of lysosomal lipid accumulation, we also found that δ-tocopherol reduces pathological phenotypes in patient fibroblasts from other lysosomal storage diseases, including NPC2, Batten (ceroid lipofuscinosis, neuronal 2, CLN2), Fabry, Farber, Niemann-Pick disease type A, Sanfilippo type B (mucopolysaccharidosis type IIIB, MPSIIIB), and Tay-Sachs. Our data suggest that regulated exocytosis may represent a potential therapeutic target for reduction of lysosomal storage in this class of diseases.


Assuntos
Sinalização do Cálcio/efeitos dos fármacos , Colesterol/metabolismo , Metabolismo dos Lipídeos/efeitos dos fármacos , Lisossomos/metabolismo , Doença de Niemann-Pick Tipo C/metabolismo , Tocoferóis/farmacologia , Doença de Wolman/metabolismo , Animais , Cálcio/metabolismo , Linhagem Celular , Cricetinae , Exocitose/efeitos dos fármacos , Humanos , Lisossomos/patologia , Doença de Niemann-Pick Tipo C/patologia , Tripeptidil-Peptidase 1 , Doença de Wolman/patologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...