STEM-03. CONSUMPTION OF A HIGH-FAT DIET INHIBITS THE TUMOR SUPPRESSIVE ACTIVITY OF HYDROGEN SULFIDE, DRIVING CANCER STEM CELL ENRICHMENT AND DISEASE AGGRESSION IN GLIOBLASTOMA

Abstract Glioblastoma (GBM) remains among the deadliest of human malignancies. Effective disease management is lacking due in part to the emergence of the cancer stem cell (CSC) phenotype. The tumor cell extrinsic, environmental, and lifestyle factors that result in CSC enrichment are not well under...

Full description

Saved in:
Bibliographic Details
Published inNeuro-oncology (Charlottesville, Va.) Vol. 21; no. Supplement_6; p. vi234
Main Authors Silver, Daniel, Roversi, Gustavo, Bithi, Nazmin, Troike, Katie, Neumann, Chase, Mark Brown, J, Hine, Christopher, Lathia, Justin
Format Journal Article
LanguageEnglish
Published US Oxford University Press 11.11.2019
Subjects
Online AccessGet full text

Cover

Loading…
More Information
Summary:Abstract Glioblastoma (GBM) remains among the deadliest of human malignancies. Effective disease management is lacking due in part to the emergence of the cancer stem cell (CSC) phenotype. The tumor cell extrinsic, environmental, and lifestyle factors that result in CSC enrichment are not well understood. Alongside other pathological features, the CSC state endows populations of tumor cells with a fluid metabolic profile that enables utilization of multiple nutrition sources. Therefore, to test the impact of diet on CSC enrichment, we interrogated disease progression in tumor-bearing mice fed either a high-fat diet (HFD), similar to the Western Pattern diet or a control low-fat diet. Compared to controls, HFD-consumption resulted in the presentation of a hyper-aggressive disease phenotype with truncated survival and tumors markedly enriched in tumor-initiating SOX2+ CSCs. To understand the underlying mechanism driving this finding, we examined tumors for the diet-regulated metabolite hydrogen sulfide (H2S). H2S is an endogenously produced bio-active gasotransmitter similar to nitric oxide. It functions principally through protein S-sulfhydration to regulate a variety of cellular programs including mitochondrial function, stress signaling and metabolism. While there is exceedingly limited information on H2S and GBM, its HFD-driven suppression has been reported in other organ systems. We discovered a significant reduction in H2S synthesis resulting from HFD-consumption in the brain of the mouse and a striking decrease in protein S-sulfhydration in human GBM tumor tissue when compared to non-cancerous control brain tissue. We demonstrated that chemical inhibition of H2S synthesis resulted in increased tumor cell viability whereas exposure to chemical H2S donors led to pronounced cell death of cultured mouse and human GBM cells. These data demonstrate for the first time, that H2S serves as a tumor suppressor for GBM. Moreover, the diet-driven suppression of H2S helps explain the hyper-aggressive in vivo phenotype that presents in response to HFD-consumption.
ISSN:1522-8517
1523-5866
DOI:10.1093/neuonc/noz175.977