Pancreatic ductal adenocarcinoma is virtually invariably a fatal disease, and is characterized by invasive and metastatic progression, as well as a striking resistance to conventional therapeutic approaches. Early pancreatic non-invasive lesions, known as pancreatic intraepithelial neoplasia (PanIN) are believed to represent initiating lesions in the generation of PDAC. Importantly, PanINs exhibit very limited proliferation, both in human pancreas and in mouse models of cancer, despite the activation of the mitogenic Ras pathway. The precise molecular pathways that restrict the progression of early PanIN lesions to more aggressive lesions remain for the most part elusive. Identifying and targeting these pathways is likely to represent new therapeutic opportunities for the treatment of pancreatic cancer. Using genetically engineered mice and cells, we have recently demonstrated that the histone deacetylase (HDAC)-associated Sin3B protein is required for cell cycle withdrawal and senescence induced by expression of oncogenic Ras. Importantly, we have demonstrated that Sin3B protein levels strongly increase in early PanIN lesions compared to normal pancreas, but decrease as the tumor progresses. These observations strongly suggest that Ras-driven cell cycle withdrawal, mediated by Sin3B-dependent repression of pro-proliferative genes, halts the progression from premalignant to invasive pancreatic lesions in vivo. In the proposed study, we will examine the molecular mechanisms underlying the lack of proliferation observed in early stage PanINs and its relationship with oncogenic Ras signaling. We will take advantage of the conditional Sin3B allele we have generated in the mouse to assess the contribution of cell cycle exit and senescence as a tumor suppressor mechanism in the pancreas, using physiologically relevant cellular systems and mouse models. Specifically, we propose to: test the hypothesis that a Sin3B-dependent cell cycle withdrawal restricts Ras-induced pancreatic-tumor progression in vivo (Aim 1);and to identify the molecular pathways engaged by Ras activation in pancreatic ductal cells, in a Sin3B-dependent manner (Aim 2). To do so, we will use a physiologically relevant primary ductal cell culture system, and genetically engineered mouse models of pancreatic cancer.

Public Health Relevance

Pancreatic ductal adenocarcinoma is virtually invariably a fatal disease, and is characterized by invasive and metastatic progression, as well as a striking resistance to conventional therapeutic approaches. Early pancreatic non-invasive lesions, known as pancreatic intraepithelial neoplasia (PanIN) are believed to represent initiating lesions in the generation of PDAC. Understanding the molecular events that allow the progression from PanINS to full cancer would provide new therapeutic opportunities in the treatment of pancreatic cancer.

Agency
National Institute of Health (NIH)
Institute
National Cancer Institute (NCI)
Type
Exploratory/Developmental Grants (R21)
Project #
3R21CA155736-02S1
Application #
8761364
Study Section
Tumor Cell Biology Study Section (TCB)
Program Officer
Hildesheim, Jeffrey
Project Start
2013-12-16
Project End
2015-12-15
Budget Start
2013-12-16
Budget End
2015-12-15
Support Year
2
Fiscal Year
2014
Total Cost
$199,078
Indirect Cost
$81,628
Name
New York University
Department
Pharmacology
Type
Schools of Medicine
DUNS #
121911077
City
New York
State
NY
Country
United States
Zip Code
10016
Cantor, David J; David, Gregory (2017) The chromatin-associated Sin3B protein is required for hematopoietic stem cell functions in mice. Blood 129:60-70
Graveline, Richard; Marcinkiewicz, Katarzyna; Choi, Seyun et al. (2017) The Chromatin-Associated Phf12 Protein Maintains Nucleolar Integrity and Prevents Premature Cellular Senescence. Mol Cell Biol 37:
Bainor, Anthony J; Deng, Fang-Ming; Wang, Yu et al. (2017) Chromatin-Associated Protein SIN3B Prevents Prostate Cancer Progression by Inducing Senescence. Cancer Res 77:5339-5348
Cantor, David J; David, Gregory (2017) The potential of targeting Sin3B and its associated complexes for cancer therapy. Expert Opin Ther Targets 21:1051-1061
Porciuncula, A; Hajdu, C; David, G (2016) The Dual Role of Senescence in Pancreatic Ductal Adenocarcinoma. Adv Cancer Res 131:1-20
DiMauro, Teresa; Cantor, David J; Bainor, Anthony J et al. (2015) Transcriptional repression of Sin3B by Bmi-1 prevents cellular senescence and is relieved by oncogene activation. Oncogene 34:4011-4017
Cantor, David J; David, Gregory (2014) SIN3B, the SASP, and pancreatic cancer. Mol Cell Oncol 1:e969167
Rielland, Maïté; Cantor, David J; Graveline, Richard et al. (2014) Senescence-associated SIN3B promotes inflammation and pancreatic cancer progression. J Clin Invest 124:2125-35
David, Gregory (2012) Regulation of oncogene-induced cell cycle exit and senescence by chromatin modifiers. Cancer Biol Ther 13:992-1000
Pellegrino, Jessica; Castrillon, Diego H; David, Gregory (2012) Chromatin associated Sin3A is essential for male germ cell lineage in the mouse. Dev Biol 369:349-55