학술논문

Intermediate filament dysregulation and astrocytopathy in the human disease model of KLHL16 mutation in giant axonal neuropathy (GAN).
Document Type
Author
Battaglia R; Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill.; Faridounnia M; Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill.; Beltran A; Department of Genetics, University of North Carolina at Chapel Hill.; Robinson J; Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill.; Kinghorn K; Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill.; Ezzell JA; Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill.; Bharucha-Goebel D; National Institute of Neurological Diseases and Stroke, Bethesda, MD.; Bonnemann C; National Institute of Neurological Diseases and Stroke, Bethesda, MD.; Hooper JE; Department of Pathology, Stanford University, Palo Alto, CA.; Opal P; Departments of Neurology and Cell and Developmental Biology, Northwestern University, Chicago, IL.; Bouldin TW; Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill.; Armao D; Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill.; Department of Radiology, University of North Carolina at Chapel Hill.; Snider N; Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill.
Source
Country of Publication: United States NLM ID: 101680187 Publication Model: Electronic Cited Medium: Internet NLM ISO Abbreviation: bioRxiv Subsets: PubMed not MEDLINE
Subject
Language
English
Abstract
Giant Axonal Neuropathy (GAN) is a pediatric neurodegenerative disease caused by KLHL16 mutations. KLHL16 encodes gigaxonin, a regulator of intermediate filament (IF) protein turnover. Previous neuropathological studies and our own examination of postmortem GAN brain tissue in the current study revealed astrocyte involvement in GAN. To study the underlying mechanisms, we reprogrammed skin fibroblasts from seven GAN patients carrying different KLHL16 mutations to iPSCs. Isogenic controls with restored IF phenotypes were derived via CRISPR/Cas9 editing of one patient carrying a homozygous missense mutation (G332R). Neural progenitor cells (NPCs), astrocytes, and brain organoids were generated through directed differentiation. All GAN iPSC lines were deficient for gigaxonin, which was restored in the isogenic control. GAN iPSCs displayed patient-specific increased vimentin expression, while GAN NPCs had decreased nestin expression compared to isogenic control. The most striking phenotypes were observed in GAN iPSC-astrocytes and brain organoids, which exhibited dense perinuclear IF accumulations and abnormal nuclear morphology. GAN patient cells with large perinuclear vimentin aggregates accumulated nuclear KLHL16 mRNA. In over-expression studies, GFAP oligomerization and perinuclear aggregation were potentiated in the presence of vimentin. As an early effector of KLHL16 mutations, vimentin may serve as a potential therapeutic target in GAN.

Online Access