Identification of molecular alterations and signaling abnormalities that initiate and maintain the malignant phenotype in glioblastoma (GBM) is a key step in designing treatment. The NF-?B and PI3K-Akt pathways are deregulated in GBM and likely to play key roles in the pathogenesis of GBM and in resistance to treatment. NF-?B activation is a hallmark of inflammation. In this proposal, we aim to characterize the role of the death domain containing kinase receptor interacting protein (RIP, RIP1), in the biology of GBM. RIP1 activates both the NF-?B and PI3K-Akt pathways in glioma cells. In addition, RIP1 negatively regulates two key tumor suppressor signaling networks, the PTEN and the p53 pathways. RIP1 levels are increased in up to 30% of GBMs but not in Grade II-III gliomas. Furthermore, RIP1 expression confers a worse prognosis in GBM, overexpressing RIP1 induces proliferation of glioma cells and silencing RIP1 inhibits proliferation of glioma cells in an animal model of glioma. The central hypothesis of this proposal is that RIP1 plays a key role in the pathogenesis of GBM and resistance to treatment by regulating key signaling pathways in tumor cells. The broad goal of this proposal is to demonstrate experimentally that RIP1 promotes the malignant phenotype of GBM.
In Specific Aim 1, we will explore the mechanistic relationships coupling RIP1 to the proliferation of glioma cells and identify RIP1 specific effector mechanisms influencing proliferation and resistance to chemotherapy in glioma cells.
In Specific Aim 2, we will determine the mechanisms and biological significance of RIP1 mediated inhibition of tumor suppressor signaling.
In Specific Aim 3 we will benchmark the effect of RIP1 expression in glioma cells using an orthotopic animal model. This study may provide useful insights into the role of inflammation in GBM and help in the design of more effective treatments targeting inflammatory signaling pathways in GBM.

Public Health Relevance

RIP1, a protein involved in inflammation, is highly expressed in GBM and confers a poor prognosis. We investigate the mechanisms used by RIP1 to promote growth of malignant glioma. Improved understanding of RIP1 in glioma may lead to design of more rational targeted treatment.

Agency
National Institute of Health (NIH)
Institute
National Institute of Neurological Disorders and Stroke (NINDS)
Type
Research Project (R01)
Project #
5R01NS062080-04
Application #
8308027
Study Section
Clinical Neuroimmunology and Brain Tumors Study Section (CNBT)
Program Officer
Fountain, Jane W
Project Start
2009-09-29
Project End
2014-07-31
Budget Start
2012-08-01
Budget End
2014-07-31
Support Year
4
Fiscal Year
2012
Total Cost
$307,720
Indirect Cost
$111,720
Name
University of Texas Sw Medical Center Dallas
Department
Neurology
Type
Schools of Medicine
DUNS #
800771545
City
Dallas
State
TX
Country
United States
Zip Code
75390
Gong, Ke; Guo, Gao; Gerber, David E et al. (2018) TNF-driven adaptive response mediates resistance to EGFR inhibition in lung cancer. J Clin Invest 128:2500-2518
Guo, Gao; Narayan, Ram N; Horton, Lindsay et al. (2017) The Role of EGFR-Met Interactions in the Pathogenesis of Glioblastoma and Resistance to Treatment. Curr Cancer Drug Targets 17:297-302
Guo, Gao; Gong, Ke; Habib, Amyn A (2017) Analysis of Constitutive EGFR Signaling Regulating IRF3 Transcriptional Activity in Cancer Cells. Methods Mol Biol 1652:183-189
Guo, Gao; Gong, Ke; Ali, Sonia et al. (2017) A TNF-JNK-Axl-ERK signaling axis mediates primary resistance to EGFR inhibition in glioblastoma. Nat Neurosci 20:1074-1084
Li, L; Puliyappadamba, V T; Chakraborty, S et al. (2015) EGFR wild type antagonizes EGFRvIII-mediated activation of Met in glioblastoma. Oncogene 34:129-134
Guo, Gao; Gong, Ke; Wohlfeld, Bryan et al. (2015) Ligand-Independent EGFR Signaling. Cancer Res 75:3436-41
Camacho, C V; Todorova, P K; Hardebeck, M C et al. (2015) DNA double-strand breaks cooperate with loss of Ink4 and Arf tumor suppressors to generate glioblastomas with frequent Met amplification. Oncogene 34:1064-72
Gil del Alcazar, Carlos Rodrigo; Hardebeck, Molly Catherine; Mukherjee, Bipasha et al. (2014) Inhibition of DNA double-strand break repair by the dual PI3K/mTOR inhibitor NVP-BEZ235 as a strategy for radiosensitization of glioblastoma. Clin Cancer Res 20:1235-48
Hatanpaa, Kimmo J; Hu, Tianshen; Vemireddy, Vamsidhara et al. (2014) High expression of the stem cell marker nestin is an adverse prognostic factor in WHO grade II-III astrocytomas and oligoastrocytomas. J Neurooncol 117:183-189
Li, L; Chakraborty, S; Yang, C-R et al. (2014) An EGFR wild type-EGFRvIII-HB-EGF feed-forward loop regulates the activation of EGFRvIII. Oncogene 33:4253-64

Showing the most recent 10 out of 17 publications