Abstract

Due to the numerous kinases in the cell, many with overlapping substrates, it is difficult to find novel substrates for a specific kinase. To identify novel substrates of cAMP-dependent protein kinase (PKA), the PKA catalytic subunit was engineered to accept bulky N(6)-substituted ATP analogs, using a chemical genetics approach initially pioneered with v-Src (1). Methionine 120 was mutated to glycine in the ATP-binding pocket of the catalytic subunit. To express the stable mutant C-subunit in Escherichia coli required co-expression with PDK1. This mutant protein was active and fully phosphorylated on Thr(197) and Ser(338). Based on its kinetic properties, the engineered C-subunit preferred N(6)(benzyl)-ATP and N(6)(phenethyl)-ATP over other ATP analogs, but still retained a 30 microm K(m) for ATP. This mutant recombinant C-subunit was used to identify three novel PKA substrates. One protein, a novel mitochondrial ChChd protein, ChChd3, was identified, suggesting that PKA may regulate mitochondria proteins.

Highlights

  • Whether identified substrates are modified by the kinase directly or indirectly via an intermediary kinase is to engineer the ATP-binding pocket of the kinase in question to accept a bulky ATP analog that cannot be used by the wild type kinase

  • They found that this single mutation opened up a new pocket that was otherwise inaccessible and termed this residue the “gatekeeper.” The best analog, based on the modeling, was found to be nitrogen at position 6 (N6)(phenethyl)ATP and experimentally, N6(phenethyl)-ATP was preferred by the mutant but was a poor substrate for wild type v-Src

  • Based on its kinetic properties, we demonstrate a clear preference for N6(benzyl)-ATP for protein kinase (PKA)-as1, but not for wild type C-subunit

Read more

Summary

Introduction

Whether identified substrates are modified by the kinase directly or indirectly via an intermediary kinase is to engineer the ATP-binding pocket of the kinase in question to accept a bulky ATP analog that cannot be used by the wild type kinase. Once the crystal structure of hematopoietic cell kinase (Hck), a Src family tyrosine kinase was available, molecular modeling was employed to explore more fully the binding pocket of Hck to find the best analog for the single mutation [5] They found that this single mutation opened up a new pocket that was otherwise inaccessible and termed this residue the “gatekeeper.” The best analog, based on the modeling, was found to be N6(phenethyl)ATP and experimentally, N6(phenethyl)-ATP was preferred by the mutant but was a poor substrate for wild type v-Src. Substitution of the gatekeeper residue has been applied to find direct substrates of over 30 kinases to date including v-Src [6] and most recently Yersinia PKA (YpkA) [7]. ChChd is endogenously expressed in mitochondria and may play a role in metal binding similar to Cox and Cox19 [10]

Methods
Results
Conclusion
Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call