Senescence is a form of stable proliferative arrest historically associated with the exhaustion of replicative potential of cells. Activated oncogenes, such as ras, can induce senescence prematurely in young cells. Recent studies demonstrate that like apoptosis, oncogene-induced senescence is a bona fide tumor suppressing mechanism in vivo, which needs to be compromised during cancer development. However, the signaling pathways responsible for this important anti-tumorigenic response are poorly understood. Studies from our lab indicate that the p38 MAPK and its downstream substrate kinase PRAK play a key role in oncogenic ras-induced senescence and tumor suppression both in vitro and in vivo, and that PRAK is likely to be a tumor suppressor protein. Studies from the last funding period demonstrated that a multifunctional acetyltransferase Tip60 is also essential for oncogenic ras-induced senescence. Further analyses revealed a novel posttranslational modification cascade involving p38, Tip60 and PRAK, which plays an essential role in oncogenic ras-induced senescence. Upon activation by ras, p38 induces the acetyltransferase activity of Tip60 through phosphorylation of Thr158; activated Tip60, which directly interacts with PRAK, in turn induces the protein kinase activity of PRAK through acetylation of K364 in a manner that depends on phosphorylation of both Tip60 and PRAK by p38. These posttranslational modifications are critical for the pro-senescent function of Tip60 and PRAK, respectively. In the current renewal application, we propose to investigate the mechanism by which Tip60-mediated acetylation induces the activity and function of PRAK (Aims 1 and 2), and to determine the role of this novel posttranslational modification cascade in senescence induction and tumor suppression in a murine cancer model (Aim 3).
In Aim 1, we will test a hypothesis that Tip60-mediated acetylation induces PRAK activity and function by enhancing p38 docking and/or by modulating its subcellular localization, 2 mechanisms that may not be mutually exclusive.
In Aim 2, we will examine a possibility that Tip60-mediated acetylation disrupts the autoinhibitory conformation in the PRAK protein, leading to activation of PRAK, using biochemical and structural biology approaches. Finally in Aim 3, we will investigate the role of the p38-Tip60-PRAK cascade during senescence induction in vivo using the DMBA-induced skin carcinogenesis mouse model. These studies will likely reveal novel molecular mechanisms that regulate the activity and function of PRAK and other similar protein kinases, and provide new insights into signaling pathways mediating senescence and tumor suppression. Investigation of the p38-Tip60-PRAK cascade in vivo may lead to the development of novel cancer therapies targeting components of the senescence pathway.

Public Health Relevance

Studies proposed in this application are designed to investigate the mechanism of oncogene-induced senescence, an important tumor suppressive cellular response that is disrupted during human cancer development. These studies focus on a protein kinase PRAK and an acetyltransferase Tip60, both of which are likely to be tumor suppressor proteins. Results from these studies will reveal mechanisms that regulate the activity and function of PRAK and other similar protein kinases, and provide new insights into signaling pathways that mediate senescence and tumor suppression, and may lead to the development of novel cancer therapies targeting the senescence pathway.

Agency
National Institute of Health (NIH)
Institute
National Cancer Institute (NCI)
Type
Research Project (R01)
Project #
7R01CA131231-07
Application #
9214821
Study Section
Cancer Molecular Pathobiology Study Section (CAMP)
Program Officer
Hildesheim, Jeffrey
Project Start
2007-12-01
Project End
2019-07-31
Budget Start
2016-03-01
Budget End
2016-07-31
Support Year
7
Fiscal Year
2015
Total Cost
$183,238
Indirect Cost
$65,020
Name
Wake Forest University Health Sciences
Department
Biology
Type
Schools of Medicine
DUNS #
937727907
City
Winston-Salem
State
NC
Country
United States
Zip Code
27157
Su, Weijun; Hong, Lixin; Xu, Xin et al. (2018) miR-30 disrupts senescence and promotes cancer by targeting both p16INK4A and DNA damage pathways. Oncogene 37:5618-5632
Shen, Wenzhi; Xie, Junling; Zhao, Shuangtao et al. (2018) ICAM3 mediates inflammatory signaling to promote cancer cell stemness. Cancer Lett 422:29-43
Fang, Yan; Wang, Juan; Wang, Guanwen et al. (2017) Inactivation of p38 MAPK contributes to stem cell-like properties of non-small cell lung cancer. Oncotarget 8:26702-26717
Xu, Yingxi; Dong, Xiaoli; Qi, Pingping et al. (2017) Sox2 Communicates with Tregs Through CCL1 to Promote the Stemness Property of Breast Cancer Cells. Stem Cells 35:2351-2365
Guo, Chunlei; Chen, Yanan; Gao, Wenjuan et al. (2017) Liposomal Nanoparticles Carrying anti-IL6R Antibody to the Tumour Microenvironment Inhibit Metastasis in Two Molecular Subtypes of Breast Cancer Mouse Models. Theranostics 7:775-788
Zhang, Jianbo; Zhou, Chen; Jiang, Huimin et al. (2017) ZEB1 induces ER-? promoter hypermethylation and confers antiestrogen resistance in breast cancer. Cell Death Dis 8:e2732
Zhou, Chen; Jiang, Huimin; Zhang, Zhen et al. (2017) ZEB1 confers stem cell-like properties in breast cancer by targeting neurogenin-3. Oncotarget :
Shen, Wenzhi; Du, Renle; Li, Jun et al. (2016) TIFA suppresses hepatocellular carcinoma progression via MALT1-dependent and -independent signaling pathways. Signal Transduct Target Ther 1:16013
Zhang, Zhuhong; Yin, Jing; Yang, Jian et al. (2016) miR-885-5p suppresses hepatocellular carcinoma metastasis and inhibits Wnt/?-catenin signaling pathway. Oncotarget 7:75038-75051
Tschan, Mario P; Federzoni, Elena A; Haimovici, Aladin et al. (2015) Human DMTF1? antagonizes DMTF1? regulation of the p14(ARF) tumor suppressor and promotes cellular proliferation. Biochim Biophys Acta 1849:1198-208

Showing the most recent 10 out of 28 publications