Your browser doesn't support javascript.
loading
Protein kinase Cα gain-of-function variant in Alzheimer's disease displays enhanced catalysis by a mechanism that evades down-regulation.
Callender, Julia A; Yang, Yimin; Lordén, Gema; Stephenson, Natalie L; Jones, Alexander C; Brognard, John; Newton, Alexandra C.
Afiliação
  • Callender JA; Department of Pharmacology, University of California, San Diego, La Jolla, CA 92093.
  • Yang Y; Biomedical Sciences Graduate Program, University of California, San Diego, La Jolla, CA 92093.
  • Lordén G; Department of Pharmacology, University of California, San Diego, La Jolla, CA 92093.
  • Stephenson NL; Department of Pharmacology, University of California, San Diego, La Jolla, CA 92093.
  • Jones AC; Cancer Research UK Manchester Institute, The University of Manchester, Manchester 4BX M20, United Kingdom.
  • Brognard J; Biomedical Sciences Graduate Program, University of California, San Diego, La Jolla, CA 92093.
  • Newton AC; Cancer Research UK Manchester Institute, The University of Manchester, Manchester 4BX M20, United Kingdom.
Proc Natl Acad Sci U S A ; 115(24): E5497-E5505, 2018 06 12.
Article em En | MEDLINE | ID: mdl-29844158
ABSTRACT
Conventional protein kinase C (PKC) family members are reversibly activated by binding to the second messengers Ca2+ and diacylglycerol, events that break autoinhibitory constraints to allow the enzyme to adopt an active, but degradation-sensitive, conformation. Perturbing these autoinhibitory constraints, resulting in protein destabilization, is one of many mechanisms by which PKC function is lost in cancer. Here, we address how a gain-of-function germline mutation in PKCα in Alzheimer's disease (AD) enhances signaling without increasing vulnerability to down-regulation. Biochemical analyses of purified protein demonstrate that this mutation results in an ∼30% increase in the catalytic rate of the activated enzyme, with no changes in the concentrations of Ca2+ or lipid required for half-maximal activation. Molecular dynamics simulations reveal that this mutation has both localized and allosteric effects, most notably decreasing the dynamics of the C-helix, a key determinant in the catalytic turnover of kinases. Consistent with this mutation not altering autoinhibitory constraints, live-cell imaging studies reveal that the basal signaling output of PKCα-M489V is unchanged. However, the mutant enzyme in cells displays increased sensitivity to an inhibitor that is ineffective toward scaffolded PKC, suggesting the altered dynamics of the kinase domain may influence protein interactions. Finally, we show that phosphorylation of a key PKC substrate, myristoylated alanine-rich C-kinase substrate, is increased in brains of CRISPR-Cas9 genome-edited mice containing the PKCα-M489V mutation. Our results unveil how an AD-associated mutation in PKCα permits enhanced agonist-dependent signaling via a mechanism that evades the cell's homeostatic down-regulation of constitutively active PKCα.
Assuntos
Palavras-chave

Texto completo: 1 Base de dados: MEDLINE Assunto principal: Regulação para Baixo / Proteína Quinase C-alfa / Doença de Alzheimer / Mutação com Ganho de Função Idioma: En Ano de publicação: 2018 Tipo de documento: Article

Texto completo: 1 Base de dados: MEDLINE Assunto principal: Regulação para Baixo / Proteína Quinase C-alfa / Doença de Alzheimer / Mutação com Ganho de Função Idioma: En Ano de publicação: 2018 Tipo de documento: Article