Your browser doesn't support javascript.
loading
Parkinson's disease-linked D620N VPS35 knockin mice manifest tau neuropathology and dopaminergic neurodegeneration.
Chen, Xi; Kordich, Jennifer K; Williams, Erin T; Levine, Nathan; Cole-Strauss, Allyson; Marshall, Lee; Labrie, Viviane; Ma, Jiyan; Lipton, Jack W; Moore, Darren J.
Afiliação
  • Chen X; Center for Neurodegenerative Science, Van Andel Research Institute, Grand Rapids, MI 49503.
  • Kordich JK; Center for Neurodegenerative Science, Van Andel Research Institute, Grand Rapids, MI 49503.
  • Williams ET; Center for Neurodegenerative Science, Van Andel Research Institute, Grand Rapids, MI 49503.
  • Levine N; Center for Neurodegenerative Science, Van Andel Research Institute, Grand Rapids, MI 49503.
  • Cole-Strauss A; Department of Translational Science and Molecular Medicine, Michigan State University, Grand Rapids, MI 49503.
  • Marshall L; Center for Neurodegenerative Science, Van Andel Research Institute, Grand Rapids, MI 49503.
  • Labrie V; Center for Neurodegenerative Science, Van Andel Research Institute, Grand Rapids, MI 49503.
  • Ma J; Division of Psychiatry and Behavioral Medicine, College of Human Medicine, Michigan State University, Grand Rapids, MI 49503.
  • Lipton JW; Center for Neurodegenerative Science, Van Andel Research Institute, Grand Rapids, MI 49503.
  • Moore DJ; Department of Translational Science and Molecular Medicine, Michigan State University, Grand Rapids, MI 49503.
Proc Natl Acad Sci U S A ; 116(12): 5765-5774, 2019 03 19.
Article em En | MEDLINE | ID: mdl-30842285
ABSTRACT
Mutations in the vacuolar protein sorting 35 ortholog (VPS35) gene represent a cause of late-onset, autosomal dominant familial Parkinson's disease (PD). A single missense mutation, D620N, is considered pathogenic based upon its segregation with disease in multiple families with PD. At present, the mechanism(s) by which familial VPS35 mutations precipitate neurodegeneration in PD are poorly understood. Here, we employ a germline D620N VPS35 knockin (KI) mouse model of PD to formally establish the age-related pathogenic effects of the D620N mutation at physiological expression levels. Our data demonstrate that a heterozygous or homozygous D620N mutation is sufficient to reproduce key neuropathological hallmarks of PD as indicated by the progressive degeneration of nigrostriatal pathway dopaminergic neurons and widespread axonal pathology. Unexpectedly, endogenous D620N VPS35 expression induces robust tau-positive somatodendritic pathology throughout the brain as indicated by abnormal hyperphosphorylated and conformation-specific tau, which may represent an important and early feature of mutant VPS35-induced neurodegeneration in PD. In contrast, we find no evidence for α-synuclein-positive neuropathology in aged VPS35 KI mice, a hallmark of Lewy body pathology in PD. D620N VPS35 expression also fails to modify the lethal neurodegenerative phenotype of human A53T-α-synuclein transgenic mice. Finally, by crossing VPS35 KI and null mice, our data demonstrate that a single D620N VPS35 allele is sufficient for survival and early maintenance of dopaminergic neurons, indicating that the D620N VPS35 protein is fully functional. Our data raise the tantalizing possibility of a pathogenic interplay between mutant VPS35 and tau for inducing neurodegeneration in PD.
Assuntos
Palavras-chave

Texto completo: 1 Coleções: 01-internacional Base de dados: MEDLINE Assunto principal: Proteínas tau / Proteínas de Transporte Vesicular Limite: Animals Idioma: En Ano de publicação: 2019 Tipo de documento: Article

Texto completo: 1 Coleções: 01-internacional Base de dados: MEDLINE Assunto principal: Proteínas tau / Proteínas de Transporte Vesicular Limite: Animals Idioma: En Ano de publicação: 2019 Tipo de documento: Article