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.
Hum Mol Genet ; 24(4): 913-25, 2015 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-25305076

RESUMO

Although Huntington's disease is caused by the expansion of a CAG triplet repeat within the context of the 3144-amino acid huntingtin protein (HTT), studies reveal that N-terminal fragments of HTT containing the expanded PolyQ region can be produced by proteolytic processing and/or aberrant splicing. N-terminal HTT fragments are also prevalent in postmortem tissue, and expression of some of these fragments in model organisms can cause pathology. This has led to the hypothesis that N-terminal peptides may be critical modulators of disease pathology, raising the possibility that targeting aberrant splicing or proteolytic processing may present attractive therapeutic targets. However, many factors can contribute to pathology, including genetic background and differential expression of transgenes, in addition to intrinsic differences between fragments and their cellular effects. We have used Drosophila as a model system to determine the relative toxicities of different naturally occurring huntingtin fragments in a system in which genetic background, transgene expression levels and post-translational proteolytic processing can be controlled. These studies reveal that among the naturally occurring N-terminal HTT peptides, the exon 1 peptide is exceptionally pathogenic and exhibits unique structural and biophysical behaviors that do not appear to be incremental changes compared with other fragments. If this proves correct, efforts to specifically reduce the levels of exon 1 peptides or to target toxicity-influencing post-translational modifications that occur with the exon 1 context are likely to have the greatest impact on pathology.


Assuntos
Éxons , Doença de Huntington/genética , Proteínas Associadas aos Microtúbulos/genética , Amiloide/metabolismo , Animais , Animais Geneticamente Modificados , Modelos Animais de Doenças , Drosophila , Proteínas de Drosophila , Expressão Gênica , Humanos , Proteína Huntingtina , Masculino , Proteínas Associadas aos Microtúbulos/química , Proteínas do Tecido Nervoso/química , Proteínas do Tecido Nervoso/genética , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/metabolismo , Fragmentos de Peptídeos/farmacologia , Peptídeos/química , Peptídeos/genética , Peptídeos/metabolismo , Agregação Patológica de Proteínas , Domínios e Motivos de Interação entre Proteínas , Proteólise
2.
Hum Mol Genet ; 20(2): 261-70, 2011 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-20952447

RESUMO

Huntington's disease (HD) is an inherited, progressive and ultimately fatal neurodegenerative disorder that is characterized by psychiatric, cognitive and motor symptoms. Among the pathways implicated in HD are those involving mitogen-activated protein kinase signaling and particularly the Ras-extracellular signal-regulated kinase (ERK) cascade. Studies in both cells and animal models suggest that ERK activation might provide a novel therapeutic target for the treatment of HD but compounds that specifically activate ERK are few. To test the hypothesis that pharmaceutical activation of ERK might be protective for HD, a polyphenol, fisetin, which was previously shown to activate the Ras-ERK cascade, was tested in three different models of HD: PC12 cells expressing mutant Httex1 under the control of an inducible promoter, Drosophila expressing mutant Httex1 and the R6/2 mouse model of HD. The results indicate that fisetin can reduce the impact of mutant huntingtin in each of these disease models. Prompted by this observation, we determined that the related polyphenol, resveratrol, also activates ERK and is protective in HD models. Notably, although more than a dozen small molecule inhibitors of ERK activation are in clinical trials, very few small molecule activators of ERK signaling are reported. Thus, fisetin, resveratrol and related compounds might be useful for the treatment of HD by virtue of their unique ability to activate ERK.


Assuntos
MAP Quinases Reguladas por Sinal Extracelular/genética , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Flavonoides/farmacologia , Doença de Huntington/enzimologia , Fármacos Neuroprotetores/farmacologia , Estilbenos/farmacologia , Animais , Modelos Animais de Doenças , Drosophila/efeitos dos fármacos , Ativação Enzimática/efeitos dos fármacos , Flavonóis , Dosagem de Genes/efeitos dos fármacos , Regulação da Expressão Gênica/efeitos dos fármacos , Proteína Huntingtina , Proteínas Quinases JNK Ativadas por Mitógeno/genética , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Camundongos , Atividade Motora/efeitos dos fármacos , Proteínas do Tecido Nervoso/genética , Proteínas Nucleares/genética , Células PC12 , Ratos , Resveratrol , Análise de Sobrevida
3.
Hum Mol Genet ; 17(23): 3767-75, 2008 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-18762557

RESUMO

Huntington's disease (HD) is associated with transcriptional dysregulation, and multiple studies with histone deacetylase (HDAC) inhibitors suggest that global approaches for restoring transcriptional balance and appropriate protein acetylation are therapeutically promising. To determine whether more targeted approaches might be effective, we have tested the impact of all the HDACs in Drosophila on Huntingtin (Htt)-induced pathology. Among the zinc-dependent or 'classic' HDACs, we find that neurodegeneration is most sensitive to levels of Rpd3. We also find that among the NAD(+)-dependent class III deacetylases, genetic or pharmacological reduction of either Sir2 or Sirt2 provides neuroprotection to Htt-challenged animals and that even greater neuroprotection is achieved when Rpd3 and Sir2 are simultaneously reduced. Our experiments suggest that longevity promoting strategies may be distinct from those that protect against neurodegeneration in Drosophila challenged with mutant human Htt. These results highlight a novel therapeutic approach for HD in the form of Sir2 inhibition and possible combinatorial inhibition of Sir2 and Rpd3.


Assuntos
Regulação para Baixo , Proteínas de Drosophila/metabolismo , Drosophila/metabolismo , Histona Desacetilases/metabolismo , Doença de Huntington/enzimologia , Doença de Huntington/patologia , Proteínas Repressoras/metabolismo , Sirtuínas/metabolismo , Animais , Modelos Animais de Doenças , Drosophila/genética , Proteínas de Drosophila/antagonistas & inibidores , Proteínas de Drosophila/genética , Histona Desacetilase 1 , Inibidores de Histona Desacetilases , Histona Desacetilases/genética , Humanos , Doença de Huntington/tratamento farmacológico , Proteínas Repressoras/antagonistas & inibidores , Proteínas Repressoras/genética , Sirtuínas/antagonistas & inibidores , Sirtuínas/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA