Expression of the major heat shock protein Hsp72 correlates with poor prognosis in many types of cancer, suggesting that Hsp72 can provide a selective advantage to cancer cells. However, our understanding of the distinct function of Hsp72 in cancer is very limited. The objective of this research program is to fill this gap of knowledge. Recently we established that Hsp72 in fact is essential for cancer cells, since depletion of Hsp72 prevented tumor formation in xenografts and precipitated growth inhibition and senescence in many cancer cell lines, but not in untransformed epithelial cells. Furthermore, we demonstrated that Hsp72 became essential for growth because of the permanent activation of PI3K or Ras oncogenic pathways in transformed cells. We hypothesize that elevated levels of Hsp72 seen in a variety of epithelial tumors function in preventing the endogenous oncogene-induced senescence program, thus allowing cancer cells to proliferate. Therefore, many tumors become """"""""addicted"""""""" to high levels of Hsp72. The proposed project will elucidate mechanisms of the Hsp72- mediated control of the oncogene-induced senescence in cancer, using both cell culture, and animal models. A critical aspect of this work will be investigation the effects of NZ28, a novel inhibitor of the heat shock response developed by us, on tumors in animals.
In Aim 1, we will establish the role of Hsp72 in the PI3K oncogenic pathway. We will test whether endogenous mPIK3CA oncogene determines dependence on Hsp72 of cancer lines established from human tumors. Further, we will elucidate signaling pathways controlled by Hsp72 that lead to the mPIK3CA-induced senescence.
In Aim 2, we will establish the role of Hsp72 in neoplastic transformation by Her2 oncogene. Expression of Her2 oncogene in cells depleted of Hsp72 led to a paradoxical effect. Instead of transformation, these cells underwent growth arrest and senescence. Similar effect of Her2 was seen upon incubation of cells with an inhibitor of the heat shock response NZ28. Here, in both cell culture and animal experiments we will clarify how Hsp72 regulates Her2 signaling in tumors. We will investigate how Hsp72 knockout affects development of Her2-induced tumors in vivo. We will also establish whether the heat shock inhibitor NZ28 can cause tumor regression in a mouse model of Her2-positive breast tumor. Overall this program will clarify how Hsp72 controls tumor development.

Public Health Relevance

A special mechanism called cell senescence has evolved that prevents proliferation of cells upon cancer development. Therefore, cells that undergo cancer transformation must acquire mutations that allow bypassing the senescence block. Here we will investigate mechanisms of bypassing cell senescence and develop small molecules that restore the block and prevent cancer.

Agency
National Institute of Health (NIH)
Institute
National Cancer Institute (NCI)
Type
Research Project (R01)
Project #
5R01CA081244-12
Application #
7942943
Study Section
Radiation Therapeutics and Biology Study Section (RTB)
Program Officer
Prasanna, Pat G
Project Start
2000-05-01
Project End
2014-08-31
Budget Start
2010-09-01
Budget End
2011-08-31
Support Year
12
Fiscal Year
2010
Total Cost
$355,597
Indirect Cost
Name
Boston University
Department
Biochemistry
Type
Schools of Medicine
DUNS #
604483045
City
Boston
State
MA
Country
United States
Zip Code
02118
Sherman, M Y; Gabai, V L (2015) Hsp70 in cancer: back to the future. Oncogene 34:4153-61
Li, Xiaokai; Colvin, Teresa; Rauch, Jennifer N et al. (2015) Validation of the Hsp70-Bag3 protein-protein interaction as a potential therapeutic target in cancer. Mol Cancer Ther 14:642-8
Yaglom, Julia A; McFarland, Christopher; Mirny, Leonid et al. (2014) Oncogene-triggered suppression of DNA repair leads to DNA instability in cancer. Oncotarget 5:8367-78
Colvin, Teresa A; Gabai, Vladimir L; Gong, Jianlin et al. (2014) Hsp70-Bag3 interactions regulate cancer-related signaling networks. Cancer Res 74:4731-40
Gabai, Vladimir L; Meng, Le; Kim, Geunwon et al. (2012) Heat shock transcription factor Hsf1 is involved in tumor progression via regulation of hypoxia-inducible factor 1 and RNA-binding protein HuR. Mol Cell Biol 32:929-40
Kim, Geunwon; Meriin, Anatoli B; Gabai, Vladimir L et al. (2012) The heat shock transcription factor Hsf1 is downregulated in DNA damage-associated senescence, contributing to the maintenance of senescence phenotype. Aging Cell 11:617-27
Sherman, Michael Y; Meng, Le; Stampfer, Martha et al. (2011) Oncogenes induce senescence with incomplete growth arrest and suppress the DNA damage response in immortalized cells. Aging Cell 10:949-61
Meng, L; Hunt, C; Yaglom, J A et al. (2011) Heat shock protein Hsp72 plays an essential role in Her2-induced mammary tumorigenesis. Oncogene 30:2836-45
Meng, L; Gabai, V L; Sherman, M Y (2010) Heat-shock transcription factor HSF1 has a critical role in human epidermal growth factor receptor-2-induced cellular transformation and tumorigenesis. Oncogene 29:5204-13
Gabai, Vladimir L; Yaglom, Julia A; Waldman, Todd et al. (2009) Heat shock protein Hsp72 controls oncogene-induced senescence pathways in cancer cells. Mol Cell Biol 29:559-69

Showing the most recent 10 out of 12 publications