4.7 Article

Genetic deficiency or pharmacological inhibition of soluble epoxide hydrolase ameliorates high fat diet-induced pancreatic β-cell dysfunction and loss

Journal

FREE RADICAL BIOLOGY AND MEDICINE
Volume 172, Issue -, Pages 48-57

Publisher

ELSEVIER SCIENCE INC
DOI: 10.1016/j.freeradbiomed.2021.05.029

Keywords

Type 2 diabetes; beta-Cell dysfunction; Dedifferentiation; Soluble epoxide hydrolase; Epoxyeicosatrienoic acids; Oxidative stress; Pharmacological inhibition

Funding

  1. NIAAA [R21AA027633]
  2. NIH [HL121324, DK095960, 1S10RR019266]
  3. Science Translation and Innovative Research (STAIR) Grant from the UC Davis Office Of Research
  4. NIDDK [R01 DK110276, R01DK107767, U24DK097154, R01DK090492, RO1DK095359]
  5. NIEHS RIVER Award [R35ES030443]
  6. NIEHS [P42ES04699]
  7. NIFA [CAD*NTR7836H]
  8. Juvenile Diabetes Research Foundation (JDRF)
  9. [U24DK092993]

Ask authors/readers for more resources

The study identified that upregulation of soluble epoxide hydrolase (sEH) expression in beta-cells under diet-induced metabolic stress could lead to beta-cell dysfunction. Genetic deficiency of sEH enhanced glucose-stimulated insulin secretion in mice, improving systemic glucose control and reducing oxidative stress and beta-cell death. Inhibition of sEH showed potential in mitigating high fat diet-induced beta-cell loss and dedifferentiation.
Pancreatic beta-cells are crucial regulators of systemic glucose homeostasis, and their dysfunction and loss are central features in type 2 diabetes. Interventions that rectify beta-cell dysfunction and loss are essential to combat this deadly malady. In the current study, we sought to delineate the role of soluble epoxide hydrolase (sEH) in beta-cells under diet-induced metabolic stress. The expression of sEH was upregulated in murine and macaque diabetes models and islets of diabetic human patients. We postulated that hyperglycemia-induced elevation in sEH leads to a reduction in its substrates, epoxyeicosatrienoic acids (EETs), and attenuates the function of beta-cells. Genetic deficiency of sEH potentiated glucose-stimulated insulin secretion in mice, likely in a cell-autonomous manner, contributing to better systemic glucose control. Consistent with this observation, genetic and pharmacological inactivation of sEH and the treatment with EETs exhibited insulinotropic effects in isolated murine islets ex vivo. Additionally, sEH deficiency enhanced glucose sensing and metabolism with elevated ATP and cAMP concentrations. This phenotype was associated with attenuated oxidative stress and diminished beta-cell death in sEH deficient islets. Moreover, pharmacological inhibition of sEH in vivo mitigated, albeit partly, high fat diet-induced beta-cell loss and dedifferentiation. The current observations provide new insights into the role of sEH in beta-cells and information that may be leveraged for the development of a mechanism-based intervention to rectify beta-cell dysfunction and loss.

Authors

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

Reviews

Primary Rating

4.7
Not enough ratings

Secondary Ratings

Novelty
-
Significance
-
Scientific rigor
-
Rate this paper

Recommended

No Data Available
No Data Available