Protein methyltransferases orchestrate epigenetic pathways through diverse posttranslational methylation. The reaction is carried out by protein lysine methyltransferases (PKMTs) through transferring the methyl group of SAM (S-adenosyl-L- methionine) to specific lysine(s) of substrates. The errors in the process have been implicated in many cancers. Accumulated evidence indicated that epigenetic diversity requires PKMTs to methylate histones and nonhistone proteins. However, few tools are available to unambiguously profile the nonhistone targets of designated PKMTs, particularly in context of proliferating cancer cells. In addition, few PKMT inhibitors are available to disrupt PKMT functions. Such situations significantly hinder our capability to develop cancer-therapeutic strategies with the PKMTs as novel targets. Our long-term goal is to elucidate and manipulate the biological functions of PKMT for cancer diagnosis and treatment. The objective of this proposal is to develop novel technologies and apply them to identify protein targets and small-molecule inhibitors of a cancer-relevant PKMT. Knowing the targets of the PKMT will be a key step toward fully understanding the epigenetic functions of the PKMT. More importantly, the inhibitors of the PKMT can be examined for their cancer-therapeutic effects. To profile the targets of the PKMT, we envision that the enzyme can be engineered to utilize SAM analogue cofactors and thus label its targets with distinct chemical groups. The distinct modifications will then be recognized by respective reporters. Meanwhile, a novel high-throughput screening approach will be implemented to identify the inhibitors of the PKMT. The impact of our target-profiling and inhibitor-identifying methods is further strengthened by their general applicability to other PKMTs.

Public Health Relevance

This proposal is expected to advance the understanding of how disrupting protein methylation affects cancer cell proliferation, and manipulate the process pharmacologically for cancer therapy. In a broad manner, the target-profiling technology proposed here will allow the comparison of the methylation profiles in normal versus cancerous cells or in non-aggressive versus aggressive cancers. This proposal further establishes the feasibility to identify methyltransferase inhibitors as potential cancer-therapeutic reagents.

Agency
National Institute of Health (NIH)
Institute
National Institute of General Medical Sciences (NIGMS)
Type
Research Project (R01)
Project #
5R01GM096056-03
Application #
8414845
Study Section
Synthetic and Biological Chemistry A Study Section (SBCA)
Program Officer
Fabian, Miles
Project Start
2011-02-01
Project End
2015-01-31
Budget Start
2013-02-01
Budget End
2014-01-31
Support Year
3
Fiscal Year
2013
Total Cost
$331,376
Indirect Cost
$150,197
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
Quintero, Cynthia M; Laursen, Kristian B; Mongan, Nigel P et al. (2018) CARM1 (PRMT4) Acts as a Transcriptional Coactivator during Retinoic Acid-Induced Embryonic Stem Cell Differentiation. J Mol Biol 430:4168-4182
Shu, Xiao; Dai, Qing; Wu, Tong et al. (2017) N6-Allyladenosine: A New Small Molecule for RNA Labeling Identified by Mutation Assay. J Am Chem Soc 139:17213-17216
Luo, Minkui (2017) Inhibition of the Kinase Cascade Can Be Quantitative. Biochemistry 56:4443-4444
LaFave, Lindsay M; Béguelin, Wendy; Koche, Richard et al. (2016) Reply to ""Uveal melanoma cells are resistant to EZH2 inhibition regardless of BAP1 status"". Nat Med 22:578-9
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
Aldawsari, Fahad S; Aguayo-Ortiz, Rodrigo; Kapilashrami, Kanishk et al. (2016) Resveratrol-salicylate derivatives as selective DNMT3 inhibitors and anticancer agents. J Enzyme Inhib Med Chem 31:695-703
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

Showing the most recent 10 out of 34 publications