Mutant HSPB1 causes loss of translational repression by binding to PCBP1, an RNA binding protein with a possible role in neurodegenerative disease.

Details

Ressource 1Download: s40478-016-0407-3 (1).pdf (1676.31 [Ko])
State: Public
Version: author
Serval ID
serval:BIB_DDAA4510E51D
Type
Article: article from journal or magazin.
Collection
Publications
Institution
Title
Mutant HSPB1 causes loss of translational repression by binding to PCBP1, an RNA binding protein with a possible role in neurodegenerative disease.
Journal
Acta neuropathologica communications
Author(s)
Geuens T., De Winter V., Rajan N., Achsel T., Mateiu L., Almeida-Souza L., Asselbergh B., Bouhy D., Auer-Grumbach M., Bagni C., Timmerman V.
ISSN
2051-5960 (Electronic)
ISSN-L
2051-5960
Publication state
Published
Issued date
11/01/2017
Peer-reviewed
Oui
Volume
5
Number
1
Pages
5
Language
english
Notes
Publication types: Journal Article
Publication Status: epublish
Abstract
The small heat shock protein HSPB1 (Hsp27) is an ubiquitously expressed molecular chaperone able to regulate various cellular functions like actin dynamics, oxidative stress regulation and anti-apoptosis. So far disease causing mutations in HSPB1 have been associated with neurodegenerative diseases such as distal hereditary motor neuropathy, Charcot-Marie-Tooth disease and amyotrophic lateral sclerosis. Most mutations in HSPB1 target its highly conserved α-crystallin domain, while other mutations affect the C- or N-terminal regions or its promotor. Mutations inside the α-crystallin domain have been shown to enhance the chaperone activity of HSPB1 and increase the binding to client proteins. However, the HSPB1-P182L mutation, located outside and downstream of the α-crystallin domain, behaves differently. This specific HSPB1 mutation results in a severe neuropathy phenotype affecting exclusively the motor neurons of the peripheral nervous system. We identified that the HSPB1-P182L mutant protein has a specifically increased interaction with the RNA binding protein poly(C)binding protein 1 (PCBP1) and results in a reduction of its translational repressive activity. RNA immunoprecipitation followed by RNA sequencing on mouse brain lead to the identification of PCBP1 mRNA targets. These targets contain larger 3'- and 5'-UTRs than average and are enriched in an RNA motif consisting of the CTCCTCCTCCTCC consensus sequence. Interestingly, next to the clear presence of neuronal transcripts among the identified PCBP1 targets we identified known genes associated with hereditary peripheral neuropathies and hereditary spastic paraplegias. We therefore conclude that HSPB1 can mediate translational repression through interaction with an RNA binding protein further supporting its role in neurodegenerative disease.

Keywords
Animals, Brain/metabolism, Carrier Proteins/genetics, Carrier Proteins/metabolism, Charcot-Marie-Tooth Disease/genetics, Charcot-Marie-Tooth Disease/metabolism, Consensus Sequence, Fibroblasts/metabolism, Gene Expression Regulation/physiology, HEK293 Cells, HSP27 Heat-Shock Proteins/genetics, HSP27 Heat-Shock Proteins/metabolism, HeLa Cells, Heat-Shock Proteins/genetics, Heat-Shock Proteins/metabolism, Heterogeneous-Nuclear Ribonucleoproteins/genetics, Heterogeneous-Nuclear Ribonucleoproteins/metabolism, Humans, Mice, Mutant Proteins/genetics, Mutant Proteins/metabolism, Mutation, Neoplasm Proteins/genetics, Neoplasm Proteins/metabolism, Protein Binding, Protein Biosynthesis/physiology, RNA, Messenger/genetics, RNA, Messenger/metabolism, Untranslated Regions, Charcot-Marie-Tooth, Distal Hereditary Motor Neuropathy, HSPB1, PCBP1, RNA immunoprecipitation
Pubmed
Web of science
Open Access
Yes
Create date
24/01/2017 19:57
Last modification date
20/08/2019 17:02
Usage data