Exp Mol Med.  2013 Aug;45(8):e36.

LRRK2 phosphorylates Snapin and inhibits interaction of Snapin with SNAP-25

Affiliations
  • 1Institute for Brain Science and Technology, Inje University, Gaegumdong, South Korea. wseol@wmcsb.co.kr
  • 2Department of Physiology and Biomedical Sciences, Seoul National University, College of Medicine, Seoul, South Korea.
  • 3Biomembrane Plasticity Research Center, Seoul National University, College of Medicine, Seoul, South Korea.
  • 4InAm Neuroscience Research Center, Sanbon Medical Center, College of Medicine, Wonkwang University, Gunpo-Si, Gyeonggido, South Korea. sonih@wku.ac.kr
  • 5Department of Molecular and Life Sciences, Hanyang University, Ansanshi, South Korea.
  • 6Department of Neurology, Sanbon Medical Center, College of Medicine, Wonkwang University, Gunpo-Si, Gyeonggido, South Korea.

Abstract

Leucine-rich repeat kinase 2 (LRRK2) is a gene that, upon mutation, causes autosomal-dominant familial Parkinson's disease (PD). Yeast two-hybrid screening revealed that Snapin, a SNAP-25 (synaptosomal-associated protein-25) interacting protein, interacts with LRRK2. An in vitro kinase assay exhibited that Snapin is phosphorylated by LRRK2. A glutathione-S-transferase (GST) pull-down assay showed that LRRK2 may interact with Snapin via its Ras-of-complex (ROC) and N-terminal domains, with no significant difference on interaction of Snapin with LRRK2 wild type (WT) or its pathogenic mutants. Further analysis by mutation study revealed that Threonine 117 of Snapin is one of the sites phosphorylated by LRRK2. Furthermore, a Snapin T117D phosphomimetic mutant decreased its interaction with SNAP-25 in the GST pull-down assay. SNAP-25 is a component of the SNARE (Soluble NSF Attachment protein REceptor) complex and is critical for the exocytosis of synaptic vesicles. Incubation of rat brain lysate with recombinant Snapin T117D, but not WT, protein caused decreased interaction of synaptotagmin with the SNARE complex based on a co-immunoprecipitation assay. We further found that LRRK2-dependent phosphorylation of Snapin in the hippocampal neurons resulted in a decrease in the number of readily releasable vesicles and the extent of exocytotic release. Combined, these data suggest that LRRK2 may regulate neurotransmitter release via control of Snapin function by inhibitory phosphorylation.

Keyword

kinase; LRRK2; Snapin; SNAP-25; SNARE vesicle; synaptotagmin

MeSH Terms

Amino Acid Sequence
Animals
Exocytosis
Female
HEK293 Cells
Humans
Mice
Molecular Sequence Data
Mutant Proteins/metabolism
Phosphorylation
Phosphothreonine/metabolism
Protein Binding
Protein Interaction Mapping
Protein Structure, Tertiary
Protein-Serine-Threonine Kinases/*metabolism
Qa-SNARE Proteins/metabolism
Rats
Rats, Sprague-Dawley
Synaptosomal-Associated Protein 25/*metabolism
Synaptotagmins/metabolism
Vesicle-Associated Membrane Protein 2/metabolism
Vesicular Transport Proteins/chemistry/*metabolism
Mutant Proteins
Qa-SNARE Proteins
Synaptosomal-Associated Protein 25
Vesicle-Associated Membrane Protein 2
Vesicular Transport Proteins
Phosphothreonine
Synaptotagmins
Protein-Serine-Threonine Kinases
Full Text Links
  • EMM
Actions
Cited
CITED
export Copy
Close
Share
  • Twitter
  • Facebook
Similar articles
Copyright © 2024 by Korean Association of Medical Journal Editors. All rights reserved.     E-mail: koreamed@kamje.or.kr