Your browser doesn't support javascript.
loading
The molecular mechanism of eukaryotic elongation factor 2 kinase activation.
Tavares, Clint D J; Ferguson, Scarlett B; Giles, David H; Wang, Qiantao; Wellmann, Rebecca M; O'Brien, John P; Warthaka, Mangalika; Brodbelt, Jennifer S; Ren, Pengyu; Dalby, Kevin N.
Affiliation
  • Tavares CD; From the Graduate Program in Cell and Molecular Biology, the Division of Medicinal Chemistry, College of Pharmacy, clinttavares@utexas.edu.
  • Ferguson SB; the Division of Medicinal Chemistry, College of Pharmacy.
  • Giles DH; the Division of Medicinal Chemistry, College of Pharmacy.
  • Wang Q; the Division of Medicinal Chemistry, College of Pharmacy, the Department of Biomedical Engineering, Cockrell School of Engineering, and.
  • Wellmann RM; the Division of Medicinal Chemistry, College of Pharmacy.
  • O'Brien JP; the Department of Chemistry and Biochemistry, College of Natural Sciences, University of Texas, Austin, Texas 78712.
  • Warthaka M; the Division of Medicinal Chemistry, College of Pharmacy.
  • Brodbelt JS; the Department of Chemistry and Biochemistry, College of Natural Sciences, University of Texas, Austin, Texas 78712.
  • Ren P; the Department of Biomedical Engineering, Cockrell School of Engineering, and.
  • Dalby KN; From the Graduate Program in Cell and Molecular Biology, the Division of Medicinal Chemistry, College of Pharmacy, dalby@austin.utexas.edu.
J Biol Chem ; 289(34): 23901-16, 2014 Aug 22.
Article in En | MEDLINE | ID: mdl-25012662
ABSTRACT
Calmodulin (CaM)-dependent eukaryotic elongation factor 2 kinase (eEF-2K) impedes protein synthesis through phosphorylation of eukaryotic elongation factor 2 (eEF-2). It is subject to complex regulation by multiple upstream signaling pathways, through poorly described mechanisms. Precise integration of these signals is critical for eEF-2K to appropriately regulate protein translation rates. Here, an allosteric mechanism comprising two sequential conformations is described for eEF-2K activation. First, Ca(2+)/CaM binds eEF-2K with high affinity (Kd(CaM)(app) = 24 ± 5 nm) to enhance its ability to autophosphorylate Thr-348 in the regulatory loop (R-loop) by > 10(4)-fold (k(auto) = 2.6 ± 0.3 s(-1)). Subsequent binding of phospho-Thr-348 to a conserved basic pocket in the kinase domain potentially drives a conformational transition of the R-loop, which is essential for efficient substrate phosphorylation. Ca(2+)/CaM binding activates autophosphorylated eEF-2K by allosterically enhancing k(cat)(app) for peptide substrate phosphorylation by 10(3)-fold. Thr-348 autophosphorylation results in a 25-fold increase in the specificity constant (k(cat)(app)/K(m)(Pep-S) (app)), with equal contributions from k(cat)(app) and K(m)(Pep-S)(app), suggesting that peptide substrate binding is partly impeded in the unphosphorylated enzyme. In cells, Thr-348 autophosphorylation appears to control the catalytic output of active eEF-2K, contributing more than 5-fold to its ability to promote eEF-2 phosphorylation. Fundamentally, eEF-2K activation appears to be analogous to an amplifier, where output volume may be controlled by either toggling the power switch (switching on the kinase) or altering the volume control (modulating stability of the active R-loop conformation). Because upstream signaling events have the potential to modulate either allosteric step, this mechanism allows for exquisite control of eEF-2K output.
Subject(s)
Key words

Full text: 1 Collection: 01-internacional Database: MEDLINE Main subject: Elongation Factor 2 Kinase Limits: Humans Language: En Journal: J Biol Chem Year: 2014 Document type: Article

Full text: 1 Collection: 01-internacional Database: MEDLINE Main subject: Elongation Factor 2 Kinase Limits: Humans Language: En Journal: J Biol Chem Year: 2014 Document type: Article
...