Proteome-wide protein methylation is an epigenetic event that is regulated by >60 protein methyltransferases (PMTs). Because conventional approaches can only uncover a small fraction of PMT substrates without knowing PMT methylome under defined cellular settings, we have only partial insight into epigenetic role(s) of PMT in many essential biological settings. Selective perturbation of PMTs is also of great use to elucidate their role(s). While PMT- associated biology is often probed via genetic perturbation, this approach, unlike small-molecule inhibitors, lacks temporal (turn on/off), spatial (loci-specific) and dose (a range of efficacy) controls. In addition, the outcomes of genetic versus pharmacological perturbation of PMTs can be different because the former completely ablates a PMT protein whereas the latter only affects a subset of PMT's functions. However, developing selective and potent small-molecule inhibitors of PMTs is challenging. The objective of this proposal is to integrate novel chemical tools (high-quality inhibitors and bioorthogonal activity probes of PMTs) with conventional methods to examine the outcome(s) and therapeutic potential of pharmacological inhibition of a PMT. We plan to define the methylation events critical for genome replication and DNA damage responses, pharmacologically manipulate them in a temporal and dose-dependent manner, and predict its outcomes using accurate animal models.

Public Health Relevance

This proposal is expected to define the functions of protein methyltransferases (PMTs) with small-molecule probes. The revealed targets and functional roles of PMTs can be useful to elicit the essential biology associated with PMTs and present novel therapeutic strategies.

Agency
National Institute of Health (NIH)
Institute
National Institute of General Medical Sciences (NIGMS)
Type
Research Project (R01)
Project #
1R01GM120570-01
Application #
9160255
Study Section
Synthetic and Biological Chemistry B Study Section (SBCB)
Program Officer
Fabian, Miles
Project Start
2016-08-01
Project End
2020-06-30
Budget Start
2016-08-01
Budget End
2017-06-30
Support Year
1
Fiscal Year
2016
Total Cost
Indirect Cost
Name
Sloan-Kettering Institute for Cancer Research
Department
Type
DUNS #
064931884
City
New York
State
NY
Country
United States
Zip Code
10065
Luo, Minkui (2018) Chemical and Biochemical Perspectives of Protein Lysine Methylation. Chem Rev 118:6656-6705
Wu, Hong; Zheng, Weihong; Eram, Mohammad S et al. (2016) Structural basis of arginine asymmetrical dimethylation by PRMT6. Biochem J 473:3049-63
Linscott, Joshua A; Kapilashrami, Kanishk; Wang, Zhen et al. (2016) Kinetic isotope effects reveal early transition state of protein lysine methyltransferase SET8. Proc Natl Acad Sci U S A 113:E8369-E8378
Tang, Haiping; Chen, Yuling; Liu, Xiaohui et al. (2016) Downregulation of HSP60 disrupts mitochondrial proteostasis to promote tumorigenesis and progression in clear cell renal cell carcinoma. Oncotarget 7:38822-38834
Warrier, Thulasi; Kapilashrami, Kanishk; Argyrou, Argyrides et al. (2016) N-methylation of a bactericidal compound as a resistance mechanism in Mycobacterium tuberculosis. Proc Natl Acad Sci U S A 113:E4523-30
Tang, Haiping; Li, Jin; Liu, Xiaohui et al. (2016) Down-regulation of HSP60 Suppresses the Proliferation of Glioblastoma Cells via the ROS/AMPK/mTOR Pathway. Sci Rep 6:28388
Butler, Kyle V; Ma, Anqi; Yu, Wenyu et al. (2016) Structure-Based Design of a Covalent Inhibitor of the SET Domain-Containing Protein 8 (SETD8) Lysine Methyltransferase. J Med Chem 59:9881-9889
LaFave, Lindsay M; Béguelin, Wendy; Koche, Richard et al. (2015) Loss of BAP1 function leads to EZH2-dependent transformation. Nat Med 21:1344-9
Luo, Minkui (2015) Inhibitors of protein methyltransferases as chemical tools. Epigenomics 7:1327-38
Blum, Gil; Ibáñez, Glorymar; Rao, Xiangjun et al. (2014) Small-molecule inhibitors of SETD8 with cellular activity. ACS Chem Biol 9:2471-8