학술논문

Zeta Inhibitory Peptide Disrupts Electrostatic Interactions That Maintain Atypical Protein Kinase C in Its Active Conformation on the Scaffold p62*
Document Type
article
Source
Journal of Biological Chemistry. 290(36)
Subject
Biochemistry and Cell Biology
Biological Sciences
Adaptor Proteins
Signal Transducing
Amino Acid Sequence
Animals
Binding Sites
Binding
Competitive
Blotting
Western
COS Cells
Chlorocebus aethiops
Fluorescence Resonance Energy Transfer
HEK293 Cells
Humans
Isoenzymes
Models
Molecular
Molecular Sequence Data
Pregnancy Proteins
Protein Binding
Protein Conformation
Protein Kinase C
Protein Structure
Tertiary
Receptors
AMPA
Sequestosome-1 Protein
Static Electricity
atypical protein kinase C
enzyme mechanism
p62 (sequestosome 1(SQSTM1))
protein-protein interaction
scaffold protein
serine/threonine protein kinase
Chemical Sciences
Medical and Health Sciences
Biochemistry & Molecular Biology
Biological sciences
Biomedical and clinical sciences
Chemical sciences
Language
Abstract
Atypical protein kinase C (aPKC) enzymes signal on protein scaffolds, yet how they are maintained in an active conformation on scaffolds is unclear. A myristoylated peptide based on the autoinhibitory pseudosubstrate fragment of the atypical PKCζ, zeta inhibitory peptide (ZIP), has been extensively used to inhibit aPKC activity; however, we have previously shown that ZIP does not inhibit the catalytic activity of aPKC isozymes in cells (Wu-Zhang, A. X., Schramm, C. L., Nabavi, S., Malinow, R., and Newton, A. C. (2012) J. Biol. Chem. 287, 12879-12885). Here we sought to identify a bona fide target of ZIP and, in so doing, unveiled a novel mechanism by which aPKCs are maintained in an active conformation on a protein scaffold. Specifically, we used protein-protein interaction network analysis, structural modeling, and protein-protein docking to predict that ZIP binds an acidic surface on the Phox and Bem1 (PB1) domain of p62, an interaction validated by peptide array analysis. Using a genetically encoded reporter for PKC activity fused to the p62 scaffold, we show that ZIP inhibits the activity of wild-type aPKC, but not a construct lacking the pseudosubstrate. These data support a model in which the pseudosubstrate of aPKCs is tethered to the acidic surface on p62, locking aPKC in an open, signaling-competent conformation. ZIP competes for binding to the acidic surface, resulting in displacement of the pseudosubstrate of aPKC and re-engagement in the substrate-binding cavity. This study not only identifies a cellular target for ZIP, but also unveils a novel mechanism by which scaffolded aPKC is maintained in an active conformation.