Your browser doesn't support javascript.
loading
AAV-Mediated Gene Augmentation Therapy Restores Critical Functions in Mutant PRPF31+/- iPSC-Derived RPE Cells.
Brydon, Elizabeth M; Bronstein, Revital; Buskin, Adriana; Lako, Majlinda; Pierce, Eric A; Fernandez-Godino, Rosario.
Affiliation
  • Brydon EM; Department of Ophthalmology, Ocular Genomics Institute, Massachusetts Eye and Ear, Harvard Medical School, Boston, MA, USA.
  • Bronstein R; Department of Ophthalmology, Ocular Genomics Institute, Massachusetts Eye and Ear, Harvard Medical School, Boston, MA, USA.
  • Buskin A; Institute of Genetic Medicine, Newcastle University, Newcastle upon Tyne, UK.
  • Lako M; Institute of Genetic Medicine, Newcastle University, Newcastle upon Tyne, UK.
  • Pierce EA; Department of Ophthalmology, Ocular Genomics Institute, Massachusetts Eye and Ear, Harvard Medical School, Boston, MA, USA.
  • Fernandez-Godino R; Department of Ophthalmology, Ocular Genomics Institute, Massachusetts Eye and Ear, Harvard Medical School, Boston, MA, USA.
Mol Ther Methods Clin Dev ; 15: 392-402, 2019 Dec 13.
Article in En | MEDLINE | ID: mdl-31890732
ABSTRACT
Retinitis pigmentosa (RP) is the most common form of inherited vision loss and is characterized by degeneration of retinal photoreceptor cells and the retinal pigment epithelium (RPE). Mutations in pre-mRNA processing factor 31 (PRPF31) cause dominant RP via haploinsufficiency with incomplete penetrance. There is good evidence that the diverse severity of this disease is a result of differing levels of expression of the wild-type allele among patients. Thus, we hypothesize that PRPF31-related RP will be amenable to treatment by adeno-associated virus (AAV)-mediated gene augmentation therapy. To test this hypothesis, we used induced pluripotent stem cells (iPSCs) with mutations in PRPF31 and differentiated them into RPE cells. The mutant PRPF31 iPSC-RPE cells recapitulate the cellular phenotype associated with the PRPF31 pathology, including defective cell structure, diminished phagocytic function, defects in ciliogenesis, and compromised barrier function. Treatment of the mutant PRPF31 iPSC-RPE cells with AAV-PRPF31 restored normal phagocytosis and cilia formation, and it partially restored structure and barrier function. These results suggest that AAV-based gene therapy targeting RPE cells holds therapeutic promise for patients with PRPF31-related RP.
Key words

Full text: 1 Collection: 01-internacional Database: MEDLINE Language: En Journal: Mol Ther Methods Clin Dev Year: 2019 Document type: Article Affiliation country: Estados Unidos

Full text: 1 Collection: 01-internacional Database: MEDLINE Language: En Journal: Mol Ther Methods Clin Dev Year: 2019 Document type: Article Affiliation country: Estados Unidos