Apoptosis is a program of cellular suicide wherein individual cells are removed from the midst of a living tissue without disturbing overall tissue architecture. The last five years have seen an exponential increase in our understanding of the cellular machinery important for execution of the apoptotic program. However, the signal transduction pathways impacting a cell's decision to live or die are much less well understood. This proposal uses both a cell-free apoptotic system derived from Xenopus eggs and intact mammalian cells to elucidate a signaling pathway involving the adaptor protein Crk and the dual-specificity cell cycle kinase, Wee1. Previous studies demonstrated an interaction between Crk and Wee1 and a requirement for both of these proteins for in vitro apoptotic signaling in egg extracts. Extensions of this work revealed a nuclear export sequence within the Crk protein and an interaction between Crk and Wee1 within the nuclei of mammalian cells. Ablation of the NES increased Crk nuclear accumulation and Crk/Weel interactions, while accelerating apoptotic induction in both mammalian cells and egg extracts. We now propose to determine what role Wee1 plays in Crk-mediated apoptotic signaling and how the Crk/Weel signal is transmitted. Specifically, we will address the role of Wee1 kinase activity/substrates in apoptotic signaling, the importance of Wee1 for Crk nuclear localization, and the recruitment of signaling partners into a Crk/Weel complex. Moreover, we will characterize additional Crk interactors (including those interacting with the Crk N-terminal SH3 domain) to further reconstruct the pathway from Crk to cell death effectors.

Agency
National Institute of Health (NIH)
Institute
National Institute of General Medical Sciences (NIGMS)
Type
Research Project (R01)
Project #
5R01GM056518-07
Application #
6755927
Study Section
Cell Development and Function Integrated Review Group (CDF)
Program Officer
Zatz, Marion M
Project Start
1997-08-01
Project End
2007-07-31
Budget Start
2004-08-01
Budget End
2005-07-31
Support Year
7
Fiscal Year
2004
Total Cost
$261,800
Indirect Cost
Name
Duke University
Department
Biochemistry
Type
Schools of Medicine
DUNS #
044387793
City
Durham
State
NC
Country
United States
Zip Code
27705
Nutt, Leta K; Margolis, Seth S; Jensen, Mette et al. (2005) Metabolic regulation of oocyte cell death through the CaMKII-mediated phosphorylation of caspase-2. Cell 123:89-103
Tashker, Jessica S; Olson, Michael; Kornbluth, Sally (2002) Post-cytochrome C protection from apoptosis conferred by a MAPK pathway in Xenopus egg extracts. Mol Biol Cell 13:393-401
Smith, Jesse J; Richardson, D Ashley; Kopf, Jan et al. (2002) Apoptotic regulation by the Crk adapter protein mediated by interactions with Wee1 and Crm1/exportin. Mol Cell Biol 22:1412-23
Olson, M; Kornbluth, S (2001) Mitochondria in apoptosis and human disease. Curr Mol Med 1:91-122
Thress, K; Song, J; Morimoto, R I et al. (2001) Reversible inhibition of Hsp70 chaperone function by Scythe and Reaper. EMBO J 20:1033-41
Smith, J J; Evans, E K; Murakami, M et al. (2000) Wee1-regulated apoptosis mediated by the crk adaptor protein in Xenopus egg extracts. J Cell Biol 151:1391-400
Kaye, F J; Modi, S; Ivanovska, I et al. (2000) A family of ubiquitin-like proteins binds the ATPase domain of Hsp70-like Stch. FEBS Lett 467:348-55
Thress, K; Evans, E K; Kornbluth, S (1999) Reaper-induced dissociation of a Scythe-sequestered cytochrome c-releasing activity. EMBO J 18:5486-93
Thress, K; Henzel, W; Shillinglaw, W et al. (1998) Scythe: a novel reaper-binding apoptotic regulator. EMBO J 17:6135-43
Kuwana, T; Smith, J J; Muzio, M et al. (1998) Apoptosis induction by caspase-8 is amplified through the mitochondrial release of cytochrome c. J Biol Chem 273:16589-94

Showing the most recent 10 out of 12 publications