Enzymes are central components of nearly all metabolic and signaling pathways in cells and tissues. The dysregulation of enzymes and their endogenous inhibitory proteins contributes to the development of several diseases, including cancer. Nonetheless, efforts to elucidate the function of specific enzymes in cancer have, to date, suffered from a lack of techniques that can assess the activity of these proteins in complex biological systems. To address this problem, we have introduced a chemical proteomic strategy termed activity-based protein profiling (ABPP) that utilizes active site-directed probes to measure changes in enzyme activity directly in native proteomes. To date, we have generated activity-based probes that target numerous enzyme classes, including proteases, lipases, histone deacetylases, and cytochrome P450s. In the previous funding period, we applied ABPP to identify several enzyme activities dysregulated in aggressive human cancer cells, including the serine proteases uPA and tPA, and the previously uncharacterized transmembrane hydrolase KIAA1363. We have shown that these enzymes play important roles in supporting the malignant properties of cancer cells, although the biochemical mechanisms for these effects remain to be fully elucidated. We have also introduced advanced """"""""tag-free"""""""" versions of ABPP that exploit the versatility of click chemistry and resolving power of mass spectrometry to enable high-content profiling of small molecule-protein interactions in living systems. Finally, we have developed a complementary proteomic platform for globally mapping the endogenous substrates of proteases termed PROTOMAP (PROtein TOpography and Migration Analysis Platform). In this competitive renewal application, we will apply our suite of ABPP and PROTOMAP technologies to test three major hypotheses of high significance to the fields of cancer and chemical biology: 1) KIAA1363 promotes cancer aggressiveness through regulation of an ether lipid signaling network, 2) dysregulated proteases contribute to cancer pathogenicity by activating and/or inactivating key signaling pathways, and 3) tag-free ABPP will offer a general and quantitative technology to map small molecule-protein interactions in living systems. We anticipate that these studies will define key enzymatic pathways that support cancer malignancy and contain new biomarkers and therapeutic targets, as well as produce methodological advances that greatly expand the scope and utility of the ABPP and PROTOMAP technologies.

Public Health Relevance

A large fraction of human enzymes remain uncharacterized in terms of their function in health and disease. We have developed advanced chemical technologies to functionally characterize enzymes directly in native biological systems. The goal of this application is to further develop and apply these technologies to identify enzymes that play important roles in cancer, which may serve as valuable new biomarkers and therapeutic targets.

Agency
National Institute of Health (NIH)
Institute
National Cancer Institute (NCI)
Type
Method to Extend Research in Time (MERIT) Award (R37)
Project #
5R37CA087660-13
Application #
8265641
Study Section
Synthetic and Biological Chemistry B Study Section (SBCB)
Program Officer
Fu, Yali
Project Start
2000-07-01
Project End
2014-02-28
Budget Start
2012-03-01
Budget End
2013-02-28
Support Year
13
Fiscal Year
2012
Total Cost
$317,378
Indirect Cost
$150,249
Name
Scripps Research Institute
Department
Type
DUNS #
781613492
City
La Jolla
State
CA
Country
United States
Zip Code
92037
Hacker, Stephan M; Backus, Keriann M; Lazear, Michael R et al. (2017) Global profiling of lysine reactivity and ligandability in the human proteome. Nat Chem 9:1181-1190
Bar-Peled, Liron; Kemper, Esther K; Suciu, Radu M et al. (2017) Chemical Proteomics Identifies Druggable Vulnerabilities in a Genetically Defined Cancer. Cell 171:696-709.e23
Whitby, Landon R; Obach, R Scott; Simon, Gabriel M et al. (2017) Quantitative Chemical Proteomic Profiling of the in Vivo Targets of Reactive Drug Metabolites. ACS Chem Biol 12:2040-2050
Guo, Chun-Jun; Chang, Fang-Yuan; Wyche, Thomas P et al. (2017) Discovery of Reactive Microbiota-Derived Metabolites that Inhibit Host Proteases. Cell 168:517-526.e18
Niessen, Sherry; Dix, Melissa M; Barbas, Sabrina et al. (2017) Proteome-wide Map of Targets of T790M-EGFR-Directed Covalent Inhibitors. Cell Chem Biol 24:1388-1400.e7
Chen, Ying-Chu; Backus, Keriann M; Merkulova, Maria et al. (2017) Covalent Modulators of the Vacuolar ATPase. J Am Chem Soc 139:639-642
Briggs, Kimberly J; Koivunen, Peppi; Cao, Shugeng et al. (2016) Paracrine Induction of HIF by Glutamate in Breast Cancer: EglN1 Senses Cysteine. Cell 166:126-39
Chen, Wentao; Dong, Jiajia; Plate, Lars et al. (2016) Arylfluorosulfates Inactivate Intracellular Lipid Binding Protein(s) through Chemoselective SuFEx Reaction with a Binding Site Tyr Residue. J Am Chem Soc 138:7353-64
Zaro, Balyn W; Whitby, Landon R; Lum, Kenneth M et al. (2016) Metabolically Labile Fumarate Esters Impart Kinetic Selectivity to Irreversible Inhibitors. J Am Chem Soc 138:15841-15844
Dugan, Amanda; Majmudar, Chinmay Y; Pricer, Rachel et al. (2016) Discovery of Enzymatic Targets of Transcriptional Activators via in Vivo Covalent Chemical Capture. J Am Chem Soc 138:12629-35

Showing the most recent 10 out of 76 publications