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

Banco de datos
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Hum Mol Genet ; 24(4): 913-25, 2015 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-25305076

RESUMEN

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.


Asunto(s)
Exones , Enfermedad de Huntington/genética , Proteínas Asociadas a Microtúbulos/genética , Amiloide/metabolismo , Animales , Animales Modificados Genéticamente , Modelos Animales de Enfermedad , Drosophila , Proteínas de Drosophila , Expresión Génica , Humanos , Proteína Huntingtina , Masculino , Proteínas Asociadas a Microtúbulos/química , Proteínas del Tejido Nervioso/química , Proteínas del Tejido Nervioso/genética , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Fragmentos de Péptidos/química , Fragmentos de Péptidos/metabolismo , Fragmentos de Péptidos/farmacología , Péptidos/química , Péptidos/genética , Péptidos/metabolismo , Agregación Patológica de Proteínas , Dominios y Motivos de Interacción de Proteínas , Proteolisis
2.
Hum Mol Genet ; 20(2): 261-70, 2011 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-20952447

RESUMEN

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.


Asunto(s)
Quinasas MAP Reguladas por Señal Extracelular/genética , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Flavonoides/farmacología , Enfermedad de Huntington/enzimología , Fármacos Neuroprotectores/farmacología , Estilbenos/farmacología , Animales , Modelos Animales de Enfermedad , Drosophila/efectos de los fármacos , Activación Enzimática/efectos de los fármacos , Flavonoles , Dosificación de Gen/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Proteína Huntingtina , Proteínas Quinasas JNK Activadas por Mitógenos/genética , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Ratones , Actividad Motora/efectos de los fármacos , Proteínas del Tejido Nervioso/genética , Proteínas Nucleares/genética , Células PC12 , Ratas , Resveratrol , Análisis de Supervivencia
3.
Hum Mol Genet ; 17(23): 3767-75, 2008 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-18762557

RESUMEN

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.


Asunto(s)
Regulación hacia Abajo , Proteínas de Drosophila/metabolismo , Drosophila/metabolismo , Histona Desacetilasas/metabolismo , Enfermedad de Huntington/enzimología , Enfermedad de Huntington/patología , Proteínas Represoras/metabolismo , Sirtuinas/metabolismo , Animales , Modelos Animales de Enfermedad , Drosophila/genética , Proteínas de Drosophila/antagonistas & inhibidores , Proteínas de Drosophila/genética , Histona Desacetilasa 1 , Inhibidores de Histona Desacetilasas , Histona Desacetilasas/genética , Humanos , Enfermedad de Huntington/tratamiento farmacológico , Proteínas Represoras/antagonistas & inhibidores , Proteínas Represoras/genética , Sirtuinas/antagonistas & inhibidores , Sirtuinas/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA