Your browser doesn't support javascript.
loading
The dynamic switch mechanism that leads to activation of LRRK2 is embedded in the DFGψ motif in the kinase domain.
Schmidt, Sven H; Knape, Matthias J; Boassa, Daniela; Mumdey, Natascha; Kornev, Alexandr P; Ellisman, Mark H; Taylor, Susan S; Herberg, Friedrich W.
Afiliação
  • Schmidt SH; Department of Biochemistry, University of Kassel, 34132 Kassel, Germany.
  • Knape MJ; Department of Biochemistry, University of Kassel, 34132 Kassel, Germany.
  • Boassa D; National Center for Microscopy and Imaging Research, University of California San Diego, La Jolla, CA 92093.
  • Mumdey N; Department of Neurosciences, University of California San Diego, La Jolla, CA 92093.
  • Kornev AP; Department of Biochemistry, University of Kassel, 34132 Kassel, Germany.
  • Ellisman MH; Department of Pharmacology, University of California San Diego, La Jolla, CA 92093.
  • Taylor SS; National Center for Microscopy and Imaging Research, University of California San Diego, La Jolla, CA 92093.
  • Herberg FW; Department of Neurosciences, University of California San Diego, La Jolla, CA 92093.
Proc Natl Acad Sci U S A ; 116(30): 14979-14988, 2019 07 23.
Article em En | MEDLINE | ID: mdl-31292254
ABSTRACT
Leucine-rich repeat kinase 2 (LRRK2) is a large multidomain protein, and LRRK2 mutants are recognized risk factors for Parkinson's disease (PD). Although the precise mechanisms that control LRRK2 regulation and function are unclear, the importance of the kinase domain is strongly implicated, since 2 of the 5 most common familial LRRK2 mutations (G2019S and I2020T) are localized to the conserved DFGψ motif in the kinase core, and kinase inhibitors are under development. Combining the concept of regulatory (R) and catalytic (C) spines with kinetic and cell-based assays, we discovered a major regulatory mechanism embedded within the kinase domain and show that the DFG motif serves as a conformational switch that drives LRRK2 activation. LRRK2 is quite unusual in that the highly conserved Phe in the DFGψ motif, which is 1 of the 4 R-spine residues, is replaced with tyrosine (DY2018GI). A Y2018F mutation creates a hyperactive phenotype similar to the familial mutation G2019S. The hydroxyl moiety of Y2018 thus serves as a "brake" that stabilizes an inactive conformation; simply removing it destroys a key hydrogen-bonding node. Y2018F, like the pathogenic mutant I2020T, spontaneously forms LRRK2-decorated microtubules in cells, while the wild type and G2019S require kinase inhibitors to form filaments. We also explored 3 different mechanisms that create kinase-dead pseudokinases, including D2017A, which further emphasizes the highly synergistic role of key hydrophobic and hydrophilic/charged residues in the assembly of active LRRK2. We thus hypothesize that LRRK2 harbors a classical protein kinase switch mechanism that drives the dynamic activation of full-length LRRK2.
Assuntos
Palavras-chave

Texto completo: 1 Coleções: 01-internacional Base de dados: MEDLINE Assunto principal: Domínio Catalítico / Simulação de Dinâmica Molecular / Serina-Treonina Proteína Quinase-2 com Repetições Ricas em Leucina Tipo de estudo: Risk_factors_studies Limite: Humans Idioma: En Ano de publicação: 2019 Tipo de documento: Article

Texto completo: 1 Coleções: 01-internacional Base de dados: MEDLINE Assunto principal: Domínio Catalítico / Simulação de Dinâmica Molecular / Serina-Treonina Proteína Quinase-2 com Repetições Ricas em Leucina Tipo de estudo: Risk_factors_studies Limite: Humans Idioma: En Ano de publicação: 2019 Tipo de documento: Article