While properly regulated levels of c-Myc are essential for normal cell growth and proliferation, aberrant overexpression and activation of c-Myc contribute to most human cancers. Thus, c-Myc level and activity must be tightly regulated during normal cell homeostasis. The rapid turnover of c-Myc is controlled by ubiquitin- dependent proteolysis. c-Myc can be ubiquitinated by the Threonine 58 phosphorylation-dependent ubiquitin ligase (E3) complex SCFFbw7 as well as various other ubiquitin E3s. Conversely, c-Myc ubiquitination can be reversed by the action of deubiquitinating enzymes (DUBs), including USP28, USP36 and USP37. Interestingly, c-Myc can also be modified by small ubiquitin-like modifiers (SUMOs). Yet the function of c-Myc SUMOylation is still unclear and how c-Myc is affected by deSUMOylation is unknown. We recently identified the SUMO protease SENP1 as a novel c-Myc deSUMOylating enzyme. SENP1 directly binds to and deSUMOylates c- Myc in cells and in vitro. Overexpression of wild-type (wt) SENP1, but not its catalytic-inactive mutant (C603S), stabilizes c-Myc and enhances c-Myc transactivation activity. Consistently, knockdown of SENP1 reduces c- Myc levels and suppresses cell proliferation. We further show that c-Myc can be co-modified by ubiquitin and SUMO and SENP1-mediated deSUMOylation reduces c-Myc ubiquitination, suggesting that SUMOylation promotes c-Myc degradation through the ubiquitin-proteasome system. In addition, SENP1 deSUMOylates USP28 whereas USP28 stabilizes SENP1 and Fbw7 reduces SENP1 levels. Thus, c-Myc levels and activity may be dynamically controlled by complex ubiquitination-SUMOylation crosstalk. SENP1 is frequently overexpressed, correlating with the high expression of c-Myc and poor patient survival, in human breast cancers. Together, these results lead to a novel hypothesis that SENP1 functions as a crucial regulator of c- Myc by deSUMOylating c-Myc. To gain further insight into the role of SENP1 in regulating c-Myc protein stability, activity and oncogenicity, we will investigate the molecular and biochemical mechanisms of the regulation of c-Myc by SENP1 in Aim 1, including how SENP1 contributes to c-Myc stabilization, how c-Myc is co-modified by SUMO and ubiquitin, and how it interplays with Fbw7 and USP28 to dynamically control c-Myc turnover. We will elucidate the role of SENP1 in c-Myc-mediated gene regulation in Aim 2 by analyzing whether SENP1 regulates c-Myc binding and turnover at target gene promoters, whether it regulates specific c- Myc target gene programs in response to growth signals, and whether SENP1 regulates the spatial localization of c-Myc in the nucleus.
In Aim 3, we will test whether SENP1 potentiates c-Myc-driven transformation and mammary tumorigenesis, whether inhibiting SENP1 suppresses c-Myc-driven tumorigenesis in vivo, and whether SENP1 inhibition is efficacious in breast cancer. Achieving these goals will provide critical insight into how c-Myc is properly regulated by dynamic SUMO modifications, how deregulation of this contributes to tumorigenesis, and whether SENP1 is a promising therapeutic target in human cancers.

Public Health Relevance

Aberrant high levels and activity of the c-Myc protein play a key role in cancer formation and progression. This study is to elucidate the molecular and cellular mechanisms underlying the regulation of c-Myc protein levels and activity by the deSUMOylating enzyme SENP1, which could yield important insight into how to target the small ubiquitin-like modifier (SUMO) pathway for cancer therapy.

Agency
National Institute of Health (NIH)
Institute
National Cancer Institute (NCI)
Type
Research Project (R01)
Project #
2R01CA186241-06A1
Application #
10058588
Study Section
Tumor Cell Biology Study Section (TCB)
Program Officer
Strasburger, Jennifer
Project Start
2015-04-01
Project End
2025-05-31
Budget Start
2020-06-01
Budget End
2021-05-31
Support Year
6
Fiscal Year
2020
Total Cost
Indirect Cost
Name
Oregon Health and Science University
Department
Other Basic Sciences
Type
Schools of Medicine
DUNS #
096997515
City
Portland
State
OR
Country
United States
Zip Code
97239
DeVine, Tiffany; Sears, Rosalie C; Dai, Mu-Shui (2018) The ubiquitin-specific protease USP36 is a conserved histone H2B deubiquitinase. Biochem Biophys Res Commun 495:2363-2368
Su, Yulong; Pelz, Carl; Huang, Tao et al. (2018) Post-translational modification localizes MYC to the nuclear pore basket to regulate a subset of target genes involved in cellular responses to environmental signals. Genes Dev 32:1398-1419
Sun, Xiao-Xin; Chen, Yingxiao; Su, Yulong et al. (2018) SUMO protease SENP1 deSUMOylates and stabilizes c-Myc. Proc Natl Acad Sci U S A 115:10983-10988
Archer, Tenley C; Ehrenberger, Tobias; Mundt, Filip et al. (2018) Proteomics, Post-translational Modifications, and Integrative Analyses Reveal Molecular Heterogeneity within Medulloblastoma Subgroups. Cancer Cell 34:396-410.e8
Chen, Yingxiao; Wang, Yue-Gang; Li, Yuhuang et al. (2017) Otub1 stabilizes MDMX and promotes its proapoptotic function at the mitochondria. Oncotarget 8:11053-11062
Janghorban, Mahnaz; Langer, Ellen M; Wang, Xiaoyan et al. (2017) The tumor suppressor phosphatase PP2A-B56? regulates stemness and promotes the initiation of malignancies in a novel murine model. PLoS One 12:e0188910
Farrell, Amy S; Joly, Meghan Morrison; Allen-Petersen, Brittany L et al. (2017) MYC regulates ductal-neuroendocrine lineage plasticity in pancreatic ductal adenocarcinoma associated with poor outcome and chemoresistance. Nat Commun 8:1728
Van Hook, Kathryn; Wang, Zhiping; Chen, Dexi et al. (2017) ?N-ASPP2, a novel isoform of the ASPP2 tumor suppressor, promotes cellular survival. Biochem Biophys Res Commun 482:1271-1277
Sun, Xiao-Xin; Sears, Rosalie C; Dai, Mu-Shui (2015) Deubiquitinating c-Myc: USP36 steps up in the nucleolus. Cell Cycle 14:3786-93
Helander, Sara; Montecchio, Meri; Pilstål, Robert et al. (2015) Pre-Anchoring of Pin1 to Unphosphorylated c-Myc in a Fuzzy Complex Regulates c-Myc Activity. Structure 23:2267-2279

Showing the most recent 10 out of 12 publications