4.8 Article

Severe consequences of a high-lipid diet include hydrogen sulfide dysfunction and enhanced aggression in glioblastoma

Journal

JOURNAL OF CLINICAL INVESTIGATION
Volume 131, Issue 17, Pages -

Publisher

AMER SOC CLINICAL INVESTIGATION INC
DOI: 10.1172/JCI138276

Keywords

-

Funding

  1. National Institutes of Health [1S10OD019972-01, 1S10RR031537-01, 1S10OD023436-01, R01 NS089641, R01 NS109742, R01 NS083629, P01 CA245705, R01 HL148352, R00 AG050777, R01 DK120679, P50 AA024333]
  2. VeloSano Bike Race
  3. Sontag Foundation Distinguished Scientist Award
  4. Case Comprehensive Cancer Center Cancer Biology Training Grant [T32 CA059366]
  5. NIH Kirschstein NRSA Fellowship [F32 CA213727]

Ask authors/readers for more resources

Research shows that consuming a high-fat diet may worsen glioblastoma (GBM), mainly by inhibiting the production of hydrogen sulfide (H2S) and increasing the enrichment of cancer stem cells (CSC). Increasing the bioavailability of H2S may help suppress the development of GBM.
Glioblastoma (GBM) remains among the deadliest of human malignancies, and the emergence of the cancer stem cell (CSC) phenotype represents a major challenge to durable treatment response. Because the environmental and lifestyle factors that impact CSC populations are not clear, we sought to understand the consequences of diet on CSC enrichment. We evaluated disease progression in mice fed an obesity-inducing high-fat diet (HFD) versus a low-fat, control diet. HFD resulted in hyperaggressive disease accompanied by CSC enrichment and shortened survival. HFD drove intracerebral accumulation of saturated fats, which inhibited the production of the cysteine metabolite and gasotransmitter, hydrogen sulfide (H2S). H2S functions principally through protein S-sulfhydration and regulates multiple programs, including bioenergetics and metabolism. Inhibition of H2S increased proliferation and chemotherapy resistance, whereas treatment with H2S donors led to death of cultured GBM cells and stasis of GBM tumors in vivo. Syngeneic GBM models and GBM patient specimens present an overall reduction in protein S-sulfhydration, primarily associated with proteins regulating cellular metabolism. These findings provide clear evidence that diet-modifiable H2S signaling serves to suppress GBM by restricting metabolic fitness, while its loss triggers CSC enrichment and disease acceleration. Interventions augmenting H2S bioavailability concurrent with GBM standard of care may improve outcomes for patients with GBM.

Authors

I am an author on this paper
Click your name to claim this paper and add it to your profile.

Reviews

Primary Rating

4.8
Not enough ratings

Secondary Ratings

Novelty
-
Significance
-
Scientific rigor
-
Rate this paper

Recommended

No Data Available
No Data Available