PUBLICATION

PRPF4 mutations cause autosomal dominant retinitis pigmentosa

Authors
Chen, X., Liu, Y., Sheng, X., Tam, P.O., Zhao, K., Chen, X., Rong, W., Liu, Y., Liu, X., Pan, X., Chen, L.J., Zhao, Q., Vollrath, D., Pang, C.P., and Zhao, C.
ID
ZDB-PUB-140317-40
Date
2014
Source
Human molecular genetics   23(11): 2926-39 (Journal)
Registered Authors
Zhao, Qingshun
Keywords
none
MeSH Terms
  • Adolescent
  • Adult
  • Aged
  • Amino Acid Sequence
  • Child
  • Down-Regulation
  • Female
  • Genes, Dominant
  • Humans
  • Male
  • Middle Aged
  • Molecular Sequence Data
  • Mutation, Missense*
  • Pedigree
  • Promoter Regions, Genetic
  • Retinitis Pigmentosa/genetics*
  • Retinitis Pigmentosa/metabolism
  • Ribonucleoprotein, U4-U6 Small Nuclear/chemistry
  • Ribonucleoprotein, U4-U6 Small Nuclear/genetics*
  • Ribonucleoprotein, U4-U6 Small Nuclear/metabolism
  • Sequence Alignment
  • Young Adult
PubMed
24419317 Full text @ Hum. Mol. Genet.
Abstract

Retinitis pigmentosa (RP), a disease characterized by progressive loss of photoreceptors, exhibits significant genetic heterogeneity. Several genes associated with U4/U6–U5 triple small nuclear ribonucleoprotein (tri-snRNP) complex of the spliceosome have been implicated in autosomal dominant RP (adRP). HPrp4, encoded by PRPF4, regulates the stability of U4/U6 di-snRNP, which is essential for continuous splicing. Here, we identified two heterozygous variants in PRPF4, including c.-114_-97del in a simplex RP patient and c.C944T (p.Pro315Leu), which co-segregates with disease phenotype in a family with adRP. Both variants were absent in 400 unrelated controls. The c.-114_-97del, predicted to affect two transcription factor binding sites, was shown to down-regulate the promoter activity of PRPF4 by a luciferase assay, and was associated with a significant reduction of PRPF4 expression in the blood cells of the patient. In fibroblasts from an affected individual with the p.Pro315Leu variant, the expression levels of several tri-snRNP components, including PRPF4 itself, were up-regulated, with altered expression pattern of SC35, a spliceosome marker. The same alterations were also observed in cells over expressing hPrp4Pro315Leu, suggesting that they arose as a compensatory response to a compromised splicing mechanism caused by hPrp4 dysfunction. Further, over expression of hPrp4Pro315Leu, but not hPrp4WT, triggered systemic deformities in wild-type zebrafish embryos with the retina primarily affected, and dramatically augmented death rates in morphant embryos, in which orthologous zebrafish prpf4 gene was silenced. We conclude that mutations of PRPF4 cause RP via haploinsufficiency and dominant-negative effects, and establish PRPF4 as a new U4/U6–U5 snRNP component associated with adRP.

Genes / Markers
Figures
Show all Figures
Expression
Phenotype
Mutations / Transgenics
Human Disease / Model
Sequence Targeting Reagents
Fish
Antibodies
Orthology
Engineered Foreign Genes
Mapping