The tumor suppressor p53 transcriptionally regulates hundreds of genes highly associated with its various cellular functions, such as cell cycle arrest, senescence and apoptosis, in response to distinct stresses. However, the kinetics of transcription of p53 targets vary dramatically regardless of their similar p53 RE sequences in most of these target genes. Often, p53 rapidly induces genes important for controlling the cell cycle, but shows delayed action on the expression of apoptotic genes. Although the biological reason for these differential actions of p53 on gene expression seems better understood, i.e., p53 might need to first rescue less genomically damaged cells through a growth arrest mechanism prior to killing them via apoptosis, the mechanisms underlying the delayed transcription of apoptotic genes by p53 remain largely unknown. In an attempt to illustrate the molecular mechanisms, we recently identified a transcriptional elongation factor TFIIS.h, whose gene is TCEA3, as a potential regulator for the expression of some p53 responsive apoptotic genes. TFIIS.h acts as a tumor suppressor in cancer cells, as its overexpression inhibits, while its knockdown promotes, growth of ovarian cancer cells. Also, our preliminary study showed that TFIIS.h is required for the transcription elongation of the bax gene, an apoptotic target of p53, but not of p21, a cell cycle target. In light of literature and our preliminary data, we hypothesize that TFIIS.h as a novel p53 target may selectively activate some of p53 responsive apoptotic genes, but not cell cycle-regulated genes, at the level of transcriptional elongation, which might account for the biochemical and molecular mechanism for why the expression of apoptotic genes, such as Bax, is often delayed in response to p53 activation. We will test this hypothesis by addressing two specific aims.
Aim 1. To identify p53 responsive TFIIS.h target genes important for apoptosis. 1.1) To validate TFIIS.h as an authentic p53 target gene: We will employ biochemical and molecular biological approaches and mouse models (p53+/+ and p53-/-) to address this. 1.2) To identify p53 responsive TFIIS.h target genes important for apoptosis. We will do so by conducting ChIP-seq and RNA-seq analyses.
Aim 2. To determine the mechanisms underlying selective regulation of transcriptional elongation of p53 responsive genes by TFIIS.h and its tumorigenic role. 2.1) To elucidate mechanisms underlying the transcriptional elongation of p53 targets by TFIIS.h. 2.2) To determine p53-dependent cellular functions of TFIIS.h, such as apoptosis, cell cycle, and senescence, by using TFIIS.h knockout cells generated by CRISPR-Cas systems. 2.3) To determine the biological effect of TFIIS.h on tumor growth. We will do so by using xenograft models. Completing these studies would not only provide new insight into the precise regulation of the kinetics of transcription of p53 target genes associated with different cellular functions, but also offer proof-of-concept evidence for selective induction of apoptosis by TFIIS.h upon p53 activation, very instrumental to the identification of novel targets for developing new therapy for chemo-resistant cancers.

Public Health Relevance

It has been known that the p53 tumor suppressor can regulate the cell cycle and apoptotic pathways by differentially regulating genes involved in these pathways, yet, it remains largely obscure how exactly p53 does so. Our unpublished studies recently revealed a transcriptional elongation factor called TFIIS.h as a new p53 target, which might play a role in regulation of p53-dependent program cell death and tumor suppression. Hence, the proposed studies in this application are aimed to understand how this protein might contribute to the differential regulations of cell cycle and apoptosis by p53, and completing the proposed studies would not only shed light onto the molecular events accounting for cancer mechanisms, but also offer profound information for potential molecular targets for future anti-cancer drug development.

Agency
National Institute of Health (NIH)
Institute
National Cancer Institute (NCI)
Type
Exploratory/Developmental Grants (R21)
Project #
5R21CA201889-02
Application #
9304032
Study Section
Special Emphasis Panel (ZCA1)
Program Officer
Watson, Joanna M
Project Start
2016-09-01
Project End
2019-02-28
Budget Start
2017-09-01
Budget End
2019-02-28
Support Year
2
Fiscal Year
2017
Total Cost
Indirect Cost
Name
Tulane University
Department
Biochemistry
Type
Schools of Medicine
DUNS #
053785812
City
New Orleans
State
LA
Country
United States
Zip Code
70118
Fang, Ziling; Cao, Bo; Liao, Jun-Ming et al. (2018) SPIN1 promotes tumorigenesis by blocking the uL18 (universal large ribosomal subunit protein 18)-MDM2-p53 pathway in human cancer. Elife 7:
Cao, Bo; Fang, Ziling; Liao, Peng et al. (2017) Cancer-mutated ribosome protein L22 (RPL22/eL22) suppresses cancer cell survival by blocking p53-MDM2 circuit. Oncotarget 8:90651-90661
Liu, Dai-Chi; Seimetz, Joseph; Lee, Kwan Young et al. (2017) Mdm2 mediates FMRP- and Gp1 mGluR-dependent protein translation and neural network activity. Hum Mol Genet 26:3895-3908
Zhang, Yiwei; Zeng, Shelya X; Hao, Qian et al. (2017) Monitoring p53 by MDM2 and MDMX is required for endocrine pancreas development and function in a spatio-temporal manner. Dev Biol 423:34-45
Liao, Peng; Zeng, Shelya X; Zhou, Xiang et al. (2017) Mutant p53 Gains Its Function via c-Myc Activation upon CDK4 Phosphorylation at Serine 249 and Consequent PIN1 Binding. Mol Cell 68:1134-1146.e6
Nguyen, Daniel; Liao, Wenjuan; Zeng, Shelya X et al. (2017) Reviving the guardian of the genome: Small molecule activators of p53. Pharmacol Ther 178:92-108
Zhang, Yu; Cao, Lan; Nguyen, Daniel et al. (2016) TP53 mutations in epithelial ovarian cancer. Transl Cancer Res 5:650-663
Chao, Tengfei; Zhou, Xiang; Cao, Bo et al. (2016) Pleckstrin homology domain-containing protein PHLDB3 supports cancer growth via a negative feedback loop involving p53. Nat Commun 7:13755
Liao, Jun-Ming; Cao, Bo; Deng, Jun et al. (2016) TFIIS.h, a new target of p53, regulates transcription efficiency of pro-apoptotic bax gene. Sci Rep 6:23542