Cardiff University | Prifysgol Caerdydd ORCA
Online Research @ Cardiff 
WelshClear Cookie - decide language by browser settings

Drug inhibition of redox factor-1 restores hypoxia-driven changes in tuberous sclerosis complex 2 deficient cells

Champion, Jesse D., Dodd, Kayleigh M., Lam, Hilaire C., Alzahrani, Mohammad A. M., Seifan, Sara, Rad, Ellie, Scourfield, David Oliver, Fishel, Melissa L., Calver, Brian L., Ager, Ann ORCID: https://orcid.org/0000-0002-5763-8908, Henske, Elizabeth P., Davies, David Mark, Kelley, Mark R. and Tee, Andrew R. ORCID: https://orcid.org/0000-0002-5577-4631 2022. Drug inhibition of redox factor-1 restores hypoxia-driven changes in tuberous sclerosis complex 2 deficient cells. Cancers 14 (24) , 6195. 10.3390/cancers14246195

[thumbnail of cancers-14-06195.pdf] PDF - Published Version
Available under License Creative Commons Attribution.

Download (4MB)

Abstract

Simple Summary: Tuberous sclerosis complex (TSC) is a genetic disease where patients are predisposed to tumors and neurological complications. Current therapies for this disease are not fully curative. We aimed to explore novel drug targets and therapies that could further benefit TSC patients. This work uncovered a novel pathway that drives disease in TSC cell models involving redox factor-1 (Ref-1). Ref-1 is a protein that turns on several key transcription factors that collectively promote tumor growth and survival through direct redox signaling. Processes regulated by Ref-1 include angiogenesis, inflammation, and metabolic transformation. Therefore, this work reveals a new drug target, where inhibitors of Ref-1 could have an additional benefit compared to current drug therapies. Abstract: Therapies with the mechanistic target of rapamycin complex 1 (mTORC1) inhibitors are not fully curative for tuberous sclerosis complex (TSC) patients. Here, we propose that some mTORC1-independent disease facets of TSC involve signaling through redox factor-1 (Ref-1). Ref-1 possesses a redox signaling activity that stimulates the transcriptional activity of STAT3, NF-kB, and HIF-1α, which are involved in inflammation, proliferation, angiogenesis, and hypoxia, respectively. Here, we demonstrate that redox signaling through Ref-1 contributes to metabolic transformation and tumor growth in TSC cell model systems. In TSC2-deficient cells, the clinically viable Ref-1 inhibitor APX3330 was effective at blocking the hyperactivity of STAT3, NF-kB, and HIF-1α. While Ref-1 inhibitors do not inhibit mTORC1, they potently block cell invasion and vasculature mimicry. Of interest, we show that cell invasion and vasculature mimicry linked to Ref-1 redox signaling are not blocked by mTORC1 inhibitors. Metabolic profiling revealed that Ref-1 inhibitors alter metabolites associated with the glutathione antioxidant pathway as well as metabolites that are heavily dysregulated in TSC2-deficient cells involved in redox homeostasis. Therefore, this work presents Ref-1 and associated redox-regulated transcription factors such as STAT3, NF-kB, and HIF-1α as potential therapeutic targets to treat TSC, where targeting these components would likely have additional benefits compared to using mTORC1 inhibitors alone.

Item Type: Article
Date Type: Published Online
Status: Published
Schools: Medicine
Additional Information: License information from Publisher: LICENSE 1: URL: https://creativecommons.org/licenses/by/4.0/, Type: open-access
Publisher: MDPI
ISSN: 2072-6694
Date of First Compliant Deposit: 6 January 2023
Date of Acceptance: 2 December 2022
Last Modified: 09 May 2023 06:21
URI: https://orca.cardiff.ac.uk/id/eprint/155462

Actions (repository staff only)

Edit Item Edit Item

Downloads

Downloads per month over past year

View more statistics