학술논문

A soluble derivative of PrPC activates cell-signaling and regulates cell physiology through LRP1 and the NMDA receptor
Document Type
article
Source
Journal of Biological Chemistry. 295(41)
Subject
Biochemistry and Cell Biology
Biomedical and Clinical Sciences
Biological Sciences
Emerging Infectious Diseases
Transmissible Spongiform Encephalopathy (TSE)
Rare Diseases
Infectious Diseases
Neurosciences
Neurological
Animals
Low Density Lipoprotein Receptor-Related Protein-1
MAP Kinase Signaling System
Neurites
PC12 Cells
PrPC Proteins
Rats
Receptors
N-Methyl-D-Aspartate
Schwann Cells
PrPC
LRP1
lipid raft
cell-signaling
neurite outgrowth
ERK1
2
Schwann cells
PC12 cells
N-methyl-d-aspartate receptor (NMDA receptor
NMDA-R)
TRK1-transforming tyrosine kinase protein
extracellular signal-regulated kinase
ERK1/2
extracellular signal–regulated kinase
Chemical Sciences
Medical and Health Sciences
Biochemistry & Molecular Biology
Biological sciences
Biomedical and clinical sciences
Chemical sciences
Language
Abstract
Cellular prion protein (PrPC) is a widely expressed glycosylphosphatidylinositol-anchored membrane protein. Scrapie prion protein is a misfolded and aggregated form of PrPC responsible for prion-induced neurodegenerative diseases. Understanding the function of the nonpathogenic PrPC monomer is an important objective. PrPC may be shed from the cell surface to generate soluble derivatives. Herein, we studied a recombinant derivative of PrPC (soluble cellular prion protein, S-PrP) that corresponds closely in sequence to a soluble form of PrPC shed from the cell surface by proteases in the A Disintegrin And Metalloprotease (ADAM) family. S-PrP activated cell-signaling in PC12 and N2a cells. TrkA was transactivated by Src family kinases and extracellular signal-regulated kinase 1/2 was activated downstream of Trk receptors. These cell-signaling events were dependent on the N-methyl-d-aspartate receptor (NMDA-R) and low-density lipoprotein receptor-related protein-1 (LRP1), which functioned as a cell-signaling receptor system in lipid rafts. Membrane-anchored PrPC and neural cell adhesion molecule were not required for S-PrP-initiated cell-signaling. S-PrP promoted PC12 cell neurite outgrowth. This response required the NMDA-R, LRP1, Src family kinases, and Trk receptors. In Schwann cells, S-PrP interacted with the LRP1/NMDA-R system to activate extracellular signal-regulated kinase 1/2 and promote cell migration. The effects of S-PrP on PC12 cell neurite outgrowth and Schwann cell migration were similar to those caused by other proteins that engage the LRP1/NMDA-R system, including activated α2-macroglobulin and tissue-type plasminogen activator. Collectively, these results demonstrate that shed forms of PrPC may exhibit important biological activities in the central nervous system and the peripheral nervous system by serving as ligands for the LRP1/NMDA-R system.