The proper regulation of gene expression in human cells is achieved by signals emanating from a distinct histone modification occurring in chromatin architecture. Studies of gene regulation mechanisms by histone modification may thus aid in the understanding and treatment of ailments caused by abnormal transcription regulation. VprBP is a nuclear protein that was originally identified on the basis of its ability to interact with HIV-1 viral protein R. Although VprBP has been studied mainly in connection with Cul4 E3 ubiquitin ligase activity, we recently discovered that VprBP is a transdominant inhibitor of the p53 tumor suppressor and counteracts p53 transactivation, apoptosis, and growth suppression functions. High-level expression of VprBP in a wide range of human tumor samples and cancer cell lines, but low to undetectable expression of VprBP in their normal counterparts, suggests that VprBP possesses oncogenic properties. Our finding that VprBP-depleted cancer cells grow very slow and do not produce tumor xenografts also supports the idea that VprBP facilitates tumorigenesis. Our studies indicated that VprBP interacts with histone H3 tails protruding from nucleosomes and that this interaction facilitates VprBP recruitment and subsequent gene silencing in cancer cells. Unexpectedly, more recent work from our laboratory uncovered the presence of kinase activity specific for threonine 120 (T120) of histone H2A in VprBP. Our functional studies demonstrated that H2A-T120 phosphorylation (H2A-T120p) is essential for VprBP-driven gene silencing in cancer cells. Based on the available evidence, VprBP is the only kinase that is responsible for H2A-T120p occurring in human cancer cells. Importantly, our development of a highly selective inhibitor to manipulate the oncogenic VprBP kinase activity sets the stage for a more detailed analysis of VprBP function in abnormal gene silencing in cancer cells. The long-term goal of the proposed research is to understand the biological processes that VprBP controls and the molecular basis of its action as a mediator of tumorigenesis. The overall objectives are to determine the mechanisms of VprBP-mediated inactivation of the genes that regulate cell proliferation and to develop a set of molecular tools for controlling the magnitude of H2A-T120p at VprBP target loci in a precise manner. Our hypothesis is that VprBP establishes and maintains the silent state of key growth regulatory genes by a two-step mechanism wherein it is recruited to target genes via interaction with gene- specific transcription factors, and phosphorylates H2A-T120 as a mark for the recruitment of additional factors involved in gene repression and cell transformation. In the first Aim, we will employ the RNA-guided CRISPR-Cas9 system in which we can manipulate H2A-T120p at specific loci, and identify the genes that are directly regulated by VprBP-mediated H2AT-120p and critical for VprBP-promoted oncogenic events. In the second Aim, we will investigate the molecular mechanisms underlying the role of H2A-T120p in maintaining the silent state of target genes by identifying and characterizing factors that selectively recognize H2A-T120p. In the third Aim, we will develop bisubstrate analogue inhibitors with higher potency toward VprBP as novel molecular tools to control H2A-T120p and block growth and proliferation of cancer cells. VprBP-mediated H2A-T120p is clearly a very important new causative mechanism for cancer development, which our lab discovered, and we have developed tools and expertise that put us in the best position to advance research on this critical subject.

Public Health Relevance

A large number of devastating human disorders result from the failure to properly control gene expression due to aberrant histone modification and chromatin regulation. Our proposal describes multiple approaches to establish VprBP as an oncogenic histone kinase and to uncover the mechanisms underlying VprBP- mediated gene repression in cancer cells. The results from this study will be useful in understanding the molecular basis of disease states that arise through misregulation of specific histone modifications as well as in inhibiting oncogenic histone modifications under cellular settings.

Agency
National Institute of Health (NIH)
Institute
National Cancer Institute (NCI)
Type
Research Project (R01)
Project #
1R01CA201561-01A1
Application #
9177706
Study Section
Cancer Molecular Pathobiology Study Section (CAMP)
Program Officer
Okano, Paul
Project Start
2016-07-15
Project End
2021-06-30
Budget Start
2016-07-15
Budget End
2017-06-30
Support Year
1
Fiscal Year
2016
Total Cost
$377,438
Indirect Cost
$148,688
Name
University of Southern California
Department
Biochemistry
Type
Schools of Medicine
DUNS #
072933393
City
Los Angeles
State
CA
Country
United States
Zip Code
90032
Kim, Kyunghwan; Shin, Yonghwan; Kim, Jinman et al. (2018) H3K27me1 is essential for MMP-9-dependent H3N-terminal tail proteolysis during osteoclastogenesis. Epigenetics Chromatin 11:23
Kim, Jin-Man; Shin, Yonghwan; Lee, Sunyoung et al. (2018) MacroH2A1.2 inhibits prostate cancer-induced osteoclastogenesis through cooperation with HP1? and H1.2. Oncogene :
Kim, Jinman; Shin, Yonghwan; Lee, Sunyoung et al. (2018) Regulation of Breast Cancer-Induced Osteoclastogenesis by MacroH2A1.2 Involving EZH2-Mediated H3K27me3. Cell Rep 24:224-237