Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 4 de 4
Filtrar
1.
Acta Neuropathol ; 135(1): 131-148, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-28780615

RESUMO

Mutations in the small heat shock protein B8 gene (HSPB8/HSP22) have been associated with distal hereditary motor neuropathy, Charcot-Marie-Tooth disease, and recently distal myopathy. It is so far not clear how mutant HSPB8 induces the neuronal and muscular phenotypes and if a common pathogenesis lies behind these diseases. Growing evidence points towards a role of HSPB8 in chaperone-associated autophagy, which has been shown to be a determinant for the clearance of poly-glutamine aggregates in neurodegenerative diseases but also for the maintenance of skeletal muscle myofibrils. To test this hypothesis and better dissect the pathomechanism of mutant HSPB8, we generated a new transgenic mouse model leading to the expression of the mutant protein (knock-in lines) or the loss-of-function (functional knock-out lines) of the endogenous protein Hspb8. While the homozygous knock-in mice developed motor deficits associated with degeneration of peripheral nerves and severe muscle atrophy corroborating patient data, homozygous knock-out mice had locomotor performances equivalent to those of wild-type animals. The distal skeletal muscles of the post-symptomatic homozygous knock-in displayed Z-disk disorganisation, granulofilamentous material accumulation along with Hspb8, αB-crystallin (HSPB5/CRYAB), and desmin aggregates. The presence of the aggregates correlated with reduced markers of effective autophagy. The sciatic nerve of the homozygous knock-in mice was characterized by low autophagy potential in pre-symptomatic and Hspb8 aggregates in post-symptomatic animals. On the other hand, the sciatic nerve of the homozygous knock-out mice presented a normal morphology and their distal muscle displayed accumulation of abnormal mitochondria but intact myofiber and Z-line organisation. Our data, therefore, suggest that toxic gain-of-function of mutant Hspb8 aggregates is a major contributor to the peripheral neuropathy and the myopathy. In addition, mutant Hspb8 induces impairments in autophagy that may aggravate the phenotype.


Assuntos
Miopatias Distais/metabolismo , Mutação com Ganho de Função , Proteínas de Choque Térmico HSP20/genética , Proteínas de Choque Térmico HSP20/metabolismo , Proteínas Musculares/genética , Proteínas Musculares/metabolismo , Miopatias Congênitas Estruturais/metabolismo , Doenças do Sistema Nervoso Periférico/metabolismo , Animais , Atrofia/metabolismo , Atrofia/patologia , Autofagia/fisiologia , Modelos Animais de Doenças , Miopatias Distais/patologia , Feminino , Proteínas de Choque Térmico , Camundongos Transgênicos , Mitocôndrias/metabolismo , Mitocôndrias/patologia , Chaperonas Moleculares , Músculo Esquelético/metabolismo , Músculo Esquelético/patologia , Miopatias Congênitas Estruturais/patologia , Nervo Isquiático/metabolismo , Nervo Isquiático/patologia
2.
Acta Neuropathol ; 126(1): 93-108, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23728742

RESUMO

Mutations in the small heat shock protein HSPB1 (HSP27) are a cause of axonal Charcot-Marie-Tooth neuropathy (CMT2F) and distal hereditary motor neuropathy. To better understand the effect of mutations in HSPB1 on the neuronal cytoskeleton, we stably transduced neuronal cells with wild-type and mutant HSPB1 and investigated axonal transport of neurofilaments (NFs). We observed that mutant HSPB1 affected the binding of NFs to the anterograde motor protein kinesin, reducing anterograde transport of NFs. These deficits were associated with an increased phosphorylation of NFs and cyclin-dependent kinase Cdk5. As Cdk5 mediates NF phosphorylation, inhibition of Cdk5/p35 restored NF phosphorylation level, as well as NF binding to kinesin in mutant HSPB1 neuronal cells. Altogether, we demonstrate that HSPB1 mutations induce hyperphosphorylation of NFs through Cdk5 and reduce anterograde transport of NFs.


Assuntos
Doença de Charcot-Marie-Tooth/genética , Quinase 5 Dependente de Ciclina/metabolismo , Proteínas de Choque Térmico HSP27/genética , Mutação/genética , Proteínas de Neurofilamentos/metabolismo , Transporte Axonal/genética , Axônios/metabolismo , Axônios/patologia , Linhagem Celular Tumoral , Quinase 5 Dependente de Ciclina/genética , Proteínas de Choque Térmico , Humanos , Imunoprecipitação , Cinesinas/metabolismo , Chaperonas Moleculares , Neuroblastoma/patologia , Fosforilação/genética , Transfecção/métodos
3.
Neuromuscul Disord ; 22(8): 699-711, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22595202

RESUMO

Missense mutations in the small heat shock protein HSPB8 cause distal hereditary motor neuropathy (dHMN) and axonal Charcot-Marie-Tooth disease (CMT2L). We previously demonstrated that, despite the ubiquitous expression of HSPB8, motor neurons appear to be predominantly affected by HSPB8 mutations. Here, we studied the effect of mutant HSPB8 in primary fibroblast cultures derived from dHMN patients' skin biopsy. In early passage cultures, we observed in all patients' fibroblasts HSPB8 protein aggregates that were not detected in control cells. After applying heat shock stress on the patients' early passage cultured cells, the protein aggregates coalesced into larger formations, while in control cells a homogenous upregulation of HSPB8 protein expression was seen. We also found a reduction in the mitochondrial membrane potential in the early passage cultures. After three months in culture, the number of cells with aggregates had become indistinguishable from that in controls and the mitochondrial membrane potential had returned to normal. These results emphasize the possible drawbacks of using patients' non-neuronal cells to study neuropathological disease mechanisms.


Assuntos
Fibroblastos/fisiologia , Proteínas de Choque Térmico/genética , Potencial da Membrana Mitocondrial/fisiologia , Doença dos Neurônios Motores/patologia , Doença dos Neurônios Motores/fisiopatologia , Mutação de Sentido Incorreto/genética , Proteínas Serina-Treonina Quinases/genética , Proteínas/metabolismo , Adulto , Idoso , Apoptose , Axônios/patologia , Biomarcadores/metabolismo , Biópsia , Células Cultivadas , Feminino , Fibroblastos/patologia , Humanos , Masculino , Pessoa de Meia-Idade , Chaperonas Moleculares , Ligação Proteica , Pele/patologia
4.
J Peripher Nerv Syst ; 17(4): 365-76, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23279337

RESUMO

The intermediate filaments called neurofilaments (NFs) are the main cytoskeleton elements in neurons. They are mainly present in the adult axonal cytoskeleton, where they are extensively phosphorylated and their phosphorylation status is pivotal for their properties and functions. Specific phosphorylation of the C-terminal domains of the large NF subunits has been implicated in radial axonal growth, NF bundling, and NF axonal transport. Many kinases and phosphatases are involved in regulating the NF phosphorylation status, and this complex interplay is of growing interest as hyperphosphorylation of NFs is a hallmark of several neurodegenerative diseases. In this review, we focus on the in vivo relevance of C-tail phosphorylation of the large NF subunits and give an overview of the kinases and phosphatases involved in regulating the phosphorylation status of the NFs.


Assuntos
Proteínas de Neurofilamentos/metabolismo , Prolina/metabolismo , Proteínas Quinases/metabolismo , Animais , Doença , Saúde , Humanos , Filamentos Intermediários/metabolismo , Degeneração Neural/metabolismo , Neurônios/metabolismo , Fosforilação
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA