The aim of Core B is to provide innovative technologies to profile RAS mutation-specific effector signaling. RAS effector signaling is complex and involves RAS interaction with a multitude (>10) of functionally diverse downstream effectors. While our current understanding of RAS effector utilization is advanced, it is also far from complete. To date, four effector families have been implicated in driving RAS-dependent cancer initiation and growth. Each effector network includes protein kinases. There is also significant crosstalk between the effector networks. Furthermore, these networks are highly dynamic, with complex feed-forward and feedback mechanisms. Classically, RAS effector signaling is profiled by evaluation of the two canonical effector pathways, the RAF-MEK-ERK mitogen-activated protein kinase cascade and the PI3K-AKT-mTOR prosurvival signaling network, using the phosphorylated state of ERK and AKT as readouts. However, it is now clear that these analyses alone fail to provide an adequate determination of RAS effector signaling. Since a major goal of this Program Project is the determination of RAS mutant-specific effector signaling, unbiased kinome-wide analyses are needed to accomplish this goal. Core B provides two innovative proteomics-based experimental platforms to accomplish this. First, Multiplexed Inhibitor Beads (MIBs) and Mass Spectroscopy (MIB/MS) analyses provide kinome-wide profiling of dynamic changes in protein kinase activity. Our preliminary studies applying MIB/MS to characterize such changes upon KRAS suppression identified protein kinases not previously known as components of RAS effector signaling, demonstrating the potential for this platform to identify novel RAS effector signaling outputs. Second, Reverse Phase Protein Array (RPPA) analyses will profile RAS-dependent changes in protein phosphorylation and activation states in cancer cell signaling networks. Additionally, a recently developed innovative advance in RPPA enabling the profiling of the activation state of interacting proteins will also be applied. The types of data generated using each experimental platform are highly complementary. We expect that together they will define novel RAS mutation- specific effector signaling networks.

Public Health Relevance

The three RAS genes comprise the most frequently mutated gene family in cancer, with highest frequencies seen in three of the top four causes of cancer deaths in the US. This proteomics core aims to better understand the complexities of RAS oncoprotein signaling to guide efforts to develop anti-RAS drugs for cancer treatment. Anti-RAS drugs are expected to make a significant impact on cancer deaths in the US.

Agency
National Institute of Health (NIH)
Institute
National Cancer Institute (NCI)
Type
Research Program Projects (P01)
Project #
5P01CA203657-02
Application #
9360646
Study Section
Special Emphasis Panel (ZCA1-RPRB-B)
Program Officer
Watson, Joanna M
Project Start
Project End
Budget Start
2017-06-01
Budget End
2018-05-31
Support Year
2
Fiscal Year
2017
Total Cost
$338,106
Indirect Cost
$82,612
Name
University of North Carolina Chapel Hill
Department
Type
Domestic Higher Education
DUNS #
608195277
City
Chapel Hill
State
NC
Country
United States
Zip Code
27599
Fu, Jingjing; Dang, Yunkun; Counter, Christopher et al. (2018) Codon usage regulates human KRAS expression at both transcriptional and translational levels. J Biol Chem 293:17929-17940
Adhikari, Hema; Counter, Christopher M (2018) Interrogating the protein interactomes of RAS isoforms identifies PIP5K1A as a KRAS-specific vulnerability. Nat Commun 9:3646
Waters, Andrew M; Der, Channing J (2018) KRAS: The Critical Driver and Therapeutic Target for Pancreatic Cancer. Cold Spring Harb Perspect Med 8:
Vaseva, Angelina V; Blake, Devon R; Gilbert, Thomas S K et al. (2018) KRAS Suppression-Induced Degradation of MYC Is Antagonized by a MEK5-ERK5 Compensatory Mechanism. Cancer Cell 34:807-822.e7
Papke, Bjoern; Der, Channing J (2017) Drugging RAS: Know the enemy. Science 355:1158-1163
Bryant, Kirsten L; Der, Channing J (2017) Mutant RAS Calms Stressed-Out Cancer Cells. Dev Cell 40:120-122
Waters, Andrew M; Ozkan-Dagliyan, Irem; Vaseva, Angelina V et al. (2017) Evaluation of the selectivity and sensitivity of isoform- and mutation-specific RAS antibodies. Sci Signal 10:
Ali, Moiez; Kaltenbrun, Erin; Anderson, Grace R et al. (2017) Codon bias imposes a targetable limitation on KRAS-driven therapeutic resistance. Nat Commun 8:15617
Yin, Guowei; Kistler, Samantha; George, Samuel D et al. (2017) A KRAS GTPase K104Q Mutant Retains Downstream Signaling by Offsetting Defects in Regulation. J Biol Chem 292:4446-4456
Huynh, Minh V; Campbell, Sharon L (2016) Getting a Handle on RAS-targeted Therapies: Cysteine Directed Inhibitors. Mini Rev Med Chem 16:383-90

Showing the most recent 10 out of 11 publications