Lung cancer is the leading cancer-related cause of death in the United States. Current treatments do not lead to a cure for most patients with this type of neoplasia. Targeted anti-tumor therapies are likely to prove more effective, but their development will require a better understanding of the signaling cascades involved. Ras oncogenes are frequently mutated in human cancers where they play an unquestionably important role in the genesis and progression of the disease. In lung adenocarcinomas, mutations in Ras are present in at least 25% of cases, suggesting that the components of Ras-related signaling pathways are promising candidates for therapeutic targets in lung cancer treatment. Ras oncogenic transformation activates NF-?B, a critical regulator of cell survival and an essential mediator of tumor progression. The atypical PKCs (aPKCs) and their adapter p62 are implicated in the control of NF-?B activation. Recent results demonstrate that genetic inactivation of the NF-?B pathway may either prevent or promote tumor development, depending on the target organ and whether or not the inflammatory response is involved. However, the effect of NF-?B inactivation in lung neoplasia has not yet been documented. Preliminary studies demonstrate that Ras reproducibly produces lung adenomas and adenocarcinomas in an inducible model of lung cancer in mice, and that p62 is required for these Ras-induced lung tumors. The evidence suggests that this is a cell-autonomous effect in that the absence of p62 severely impairs the ability of Ras to transform immortal embryo fibroblasts. In addition, Ras transformation induces p62 protein accumulation through a yet-to-be-defined mechanism. These important observations strongly indicate that p62 is necessary for Ras-induced lung tumorigenesis and cell transformation. The long-term goal of this project is to define novel signaling molecules that could be therapeutic targets in lung cancer. The overall objective of this proposal is to rigorously test the hypothesis that the p62/aPKC/NF-?B pathway is critical in Ras-induced tumor transformation, specifically in lung cancer. This will be achieved by assessing the roles and mechanism of action of these important signaling mediators in the regulation of Ras-induced transformation.
The specific aims designed to achieve this are to 1) Test the hypothesis that p62 regulates Ras-induced lung cancer in vivo through the aPKCs;2) Test the hypothesis that IKK2 or IKK1 are critical mediators of Ras-induced formation of lung tumors;3) Elucidate the cellular and molecular mechanisms of the p62/aPKC/NF-?B pathway in Ras-induced transformation and its relevance in human lung cancer. This work will increase our understanding of the mechanisms involved in the regulation of lung cancer, and in the long term will provide the knowledge necessary for the development of novel, more specific, and thus less toxic, therapies for this type of neoplasia.

Public Health Relevance

Lung cancer is the leading cancer-related cause of death in the United States, with an estimated 213,380 new cases and 160,390 deaths in 2007. Current treatments do not lead to a cure for most patients with this type of neoplasia. This research will elucidate the molecular signals implicated in the initiation of lung neoplasias. It is expected that the knowledge gained through this work will contribute to the development of new, more specific and, thus, more effective drug therapies for this type of cancer.

Agency
National Institute of Health (NIH)
Institute
National Cancer Institute (NCI)
Type
Research Project (R01)
Project #
7R01CA132847-03
Application #
8079456
Study Section
Molecular Oncogenesis Study Section (MONC)
Program Officer
Watson, Joanna M
Project Start
2009-08-01
Project End
2014-05-31
Budget Start
2011-06-01
Budget End
2012-05-31
Support Year
3
Fiscal Year
2011
Total Cost
$393,697
Indirect Cost
Name
Sanford-Burnham Medical Research Institute
Department
Type
DUNS #
020520466
City
La Jolla
State
CA
Country
United States
Zip Code
92037
Todoric, Jelena; Antonucci, Laura; Di Caro, Giuseppe et al. (2017) Stress-Activated NRF2-MDM2 Cascade Controls Neoplastic Progression in Pancreas. Cancer Cell 32:824-839.e8
Zhong, Zhenyu; Umemura, Atsushi; Sanchez-Lopez, Elsa et al. (2016) NF-?B Restricts Inflammasome Activation via Elimination of Damaged Mitochondria. Cell 164:896-910
Linares, Juan F; Duran, Angeles; Reina-Campos, Miguel et al. (2015) Amino Acid Activation of mTORC1 by a PB1-Domain-Driven Kinase Complex Cascade. Cell Rep 12:1339-52
Llado, Victoria; Nakanishi, Yuki; Duran, Angeles et al. (2015) Repression of Intestinal Stem Cell Function and Tumorigenesis through Direct Phosphorylation of ?-Catenin and Yap by PKC? Cell Rep :
Chang, Kyung Hee; Sengupta, Amitava; Nayak, Ramesh C et al. (2014) p62 is required for stem cell/progenitor retention through inhibition of IKK/NF-?B/Ccl4 signaling at the bone marrow macrophage-osteoblast niche. Cell Rep 9:2084-97
Valencia, Tania; Kim, Ji Young; Abu-Baker, Shadi et al. (2014) Metabolic reprogramming of stromal fibroblasts through p62-mTORC1 signaling promotes inflammation and tumorigenesis. Cancer Cell 26:121-135
Linares, Juan F; Duran, Angeles; Yajima, Tomoko et al. (2013) K63 polyubiquitination and activation of mTOR by the p62-TRAF6 complex in nutrient-activated cells. Mol Cell 51:283-96
Yan, Jin; Seibenhener, Michael Lamar; Calderilla-Barbosa, Luis et al. (2013) SQSTM1/p62 interacts with HDAC6 and regulates deacetylase activity. PLoS One 8:e76016
Kim, Ji Young; Valencia, Tania; Abu-Baker, Shadi et al. (2013) c-Myc phosphorylation by PKC? represses prostate tumorigenesis. Proc Natl Acad Sci U S A 110:6418-23
Ma, Li; Tao, Yongzhen; Duran, Angeles et al. (2013) Control of nutrient stress-induced metabolic reprogramming by PKC? in tumorigenesis. Cell 152:599-611

Showing the most recent 10 out of 25 publications