Abstract

Ack1 is a nonreceptor tyrosine kinase that participates in tumorigenesis, cell survival, and migration. Relatively little is known about the mechanisms that regulate Ack1 activity. Recently, four somatic missense mutations of Ack1 were identified in cancer tissue samples, but the effects on Ack1 activity, and function have not been described. These mutations occur in the N-terminal region, the C-lobe of the kinase domain, and the SH3 domain. Here, we show that the cancer-associated mutations increase Ack1 autophosphorylation in mammalian cells without affecting localization and increase Ack1 activity in immune complex kinase assays. The cancer-associated mutations potentiate the ability of Ack1 to promote proliferation and migration, suggesting that point mutation is a mechanism for Ack1 deregulation. We propose that the C-terminal Mig6 homology region (MHR) (residues 802-990) participates in inhibitory intramolecular interactions. The isolated kinase domain of Ack1 interacts directly with the MHR, and the cancer-associated E346K mutation prevents binding. Likewise, mutation of a key hydrophobic residue in the MHR (Phe(820)) prevents the MHR-kinase interaction, activates Ack1, and increases cell migration. Thus, the cancer-associated mutation E346K appears to destabilize an autoinhibited conformation of Ack1, leading to constitutively high Ack1 activity.

Highlights

  • We propose that the C-terminal Mig6 homology region (MHR) participates in inhibitory intramolecular interactions

  • Ack1 is a nonreceptor tyrosine kinase (NRTK)2 that was first isolated from a human hippocampal expression library because of its specific binding to GTP-bound Cdc42 [1]

  • Ack1 is recruited to EGFR following EGF stimulation, and overexpression of Ack1 interferes with the proper trafficking of EGFR [24]

Read more

Summary

Introduction

Ack1 is a nonreceptor tyrosine kinase (NRTK)2 that was first isolated from a human hippocampal expression library because of its specific binding to GTP-bound Cdc42 [1]. We show that the cancer-associated mutations increase Ack1 autophosphorylation in mammalian cells without affecting localization and increase Ack1 activity in immune complex kinase assays. We propose that an autoinhibitory interaction exists between the kinase domain and the C-terminal Mig6 homology region and that mutations that disrupt this interaction (such as the cancer-associated mutation E346K) activate Ack1.

Objectives
Results
Conclusion
Full Text
Paper version not known

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

Disclaimer: All third-party content on this website/platform is and will remain the property of their respective owners and is provided on "as is" basis without any warranties, express or implied. Use of third-party content does not indicate any affiliation, sponsorship with or endorsement by them. Any references to third-party content is to identify the corresponding services and shall be considered fair use under The CopyrightLaw.