Glioblastomas (GBMs) are highly lethal brain tumors with extremely poor prognosis. The treatment of GBMs remains palliative due to therapeutic resistance. GBM displays striking cellular heterogeneity with a population of tumor-initiating cell (BTICs) or GBM stem cells (GSCs) at the apex of differentiation hierarchy. We previously demonstrated that GSCs promote therapeutic resistance, tumor angiogenesis and cancer invasion, indicating that therapeutic targeting of GSCs may effectively improve GBM treatment. The stem cell-like phenotype of GSCs is maintained by a set of core transcription factors including SOX2. The protein stability of SOX2 is tightly regulated by post-translational modifications. Our preliminary study indicated that SOX2 is regulated by both ubiquitylation (Ub) and deubiquitylation (Dub) in glioma cells. SOX2 is ubiquitylated by the E3 ubiquitin ligase, Huwe1, and then targeted for degradation in non-stem glioma cells. In GSCs, SOX2 protein is stabilized through deubiquitylation mediated by the deubiquitylase, HAUSP (Herpesvirus-Associated Ubiquitin-Specific Protease, also known as USP7). HAUSP is preferentially expressed in GSCs relative to neural progenitors. Targeting HAUSP by shRNA reduced SOX2 protein, promoted cell differentiation, disrupted GSC maintenance and potently inhibited GSC tumor growth. Functional inhibition of HAUSP by a small molecule inhibitor also inhibited tumor growth in orthotopic GBM xenografts and sensitized GSCs to radiation or temozolomide (TMZ). Based on these preliminary data, we hypothesize that HAUSP-mediated deubiquitylation antagonizes Huwe1-mediated ubiquitylation to stabilize SOX2 in the maintenance of GSCs, presenting a therapeutic target strategy to disrupts GSCs and suppress GBM tumor growth. We propose the following specific aims: 1. Determine the therapeutic impact of HAUSP inhibition on suppressing GBM tumor growth. 2. Define the molecular mechanisms underlying HAUSP-mediated GSC maintenance. 3. Evaluate synergy of HAUSP inhibition and current GBM therapies. The goal of this proposal is to evaluate the therapeutic potential of HAUSP inhibition in a preclinical study. We will determine whether targeting HAUSP can serve as an effective therapeutic strategy to improve GBM treatment. The proposed studies will lay a solid foundation for the development of novel anti-GSC therapeutics to effectively improve the survival of GBM patients.

Public Health Relevance

Glioblastoma (GBM) is among the deadliest of all cancers and is highly resistant to conventional therapies, indicating that more effective therapeutics is urgently needed. We previously demonstrated that GBM stem cells (GSCs) promote therapeutic resistance and tumor growth, suggesting that targeting GSCs may effectively improve GBM treatment. GSCs are maintained by a set of core regulators including SOX2. We recently identified a critical deubiquitylating enzyme called HAUSP that stabilizes SOX2 protein to maintain the GSC phenotype. HAUSP inhibition reduced SOX2 protein, disrupted GSC maintenance, and potently inhibited tumor growth. Targeting HAUSP by a small molecule inhibitor also disrupted GSC tumor growth and sensitized GSCs to radiation or temozolomide (TMZ), suggesting that HAUSP is a potential therapeutic target. In this proposal, we will evaluate the therapeutic efficacy of targeting HAUSP in a preclinical study and determine whether HAUSP inhibition can serve as an effective strategy to improve GBM treatment. We expect that the proposed studies will lay a solid foundation for the development of novel anti-GSC specific therapeutics to improve the survival of GBM patients.

Agency
National Institute of Health (NIH)
Institute
National Cancer Institute (NCI)
Type
Research Project (R01)
Project #
5R01CA184090-05
Application #
9662787
Study Section
Developmental Therapeutics Study Section (DT)
Program Officer
Forry, Suzanne L
Project Start
2015-04-01
Project End
2021-03-31
Budget Start
2019-04-01
Budget End
2021-03-31
Support Year
5
Fiscal Year
2019
Total Cost
Indirect Cost
Name
Cleveland Clinic Lerner
Department
Other Basic Sciences
Type
Schools of Medicine
DUNS #
135781701
City
Cleveland
State
OH
Country
United States
Zip Code
44195
Man, Jianghong; Yu, Xingjiang; Huang, Haidong et al. (2018) Hypoxic Induction of Vasorin Regulates Notch1 Turnover to Maintain Glioma Stem-like Cells. Cell Stem Cell 22:104-118.e6
Wang, Xiuxing; Prager, Briana C; Wu, Qiulian et al. (2018) Reciprocal Signaling between Glioblastoma Stem Cells and Differentiated Tumor Cells Promotes Malignant Progression. Cell Stem Cell 22:514-528.e5
Shi, Yu; Ping, Yi-Fang; Zhou, Wenchao et al. (2017) Tumour-associated macrophages secrete pleiotrophin to promote PTPRZ1 signalling in glioblastoma stem cells for tumour growth. Nat Commun 8:15080
Zhou, Wenchao; Chen, Cong; Shi, Yu et al. (2017) Targeting Glioma Stem Cell-Derived Pericytes Disrupts the Blood-Tumor Barrier and Improves Chemotherapeutic Efficacy. Cell Stem Cell 21:591-603.e4
Wang, Xiuxing; Yang, Kailin; Xie, Qi et al. (2017) Purine synthesis promotes maintenance of brain tumor initiating cells in glioma. Nat Neurosci 20:661-673
Wang, Xiuxing; Huang, Zhi; Wu, Qiulian et al. (2017) MYC-Regulated Mevalonate Metabolism Maintains Brain Tumor-Initiating Cells. Cancer Res 77:4947-4960
Fang, Xiaoguang; Zhou, Wenchao; Wu, Qiulian et al. (2017) Deubiquitinase USP13 maintains glioblastoma stem cells by antagonizing FBXL14-mediated Myc ubiquitination. J Exp Med 214:245-267
Shi, Yu; Zhou, Wenchao; Cheng, Lin et al. (2017) Tetraspanin CD9 stabilizes gp130 by preventing its ubiquitin-dependent lysosomal degradation to promote STAT3 activation in glioma stem cells. Cell Death Differ 24:167-180
Jin, Xun; Kim, Leo J Y; Wu, Qiulian et al. (2017) Targeting glioma stem cells through combined BMI1 and EZH2 inhibition. Nat Med 23:1352-1361
Zhou, Wenchao; Cheng, Lin; Shi, Yu et al. (2015) Arsenic trioxide disrupts glioma stem cells via promoting PML degradation to inhibit tumor growth. Oncotarget 6:37300-15