3.9 Article

An Acetate-Specific GPCR, FFAR2, Regulates Insulin Secretion

Journal

MOLECULAR ENDOCRINOLOGY
Volume 29, Issue 7, Pages 1055-1066

Publisher

OXFORD UNIV PRESS INC
DOI: 10.1210/me.2015-1007

Keywords

-

Funding

  1. Department of Veterans Affairs, Veterans Health Administration, Office of Research and Development, Career Development Grant [1IK2BX001587-01]
  2. American Heart Association Postdoctoral Fellowship [15POST22410016]
  3. Northwestern University Program in Endocrinology, Diabetes and Hormone Action Grant [NIH T32 DK007169]
  4. North-western University Cellular and Molecular Basis of Disease Training Grant [NIH T32 GM08061]
  5. National Institutes of Health [R01 DK060581, UC4 DK104166, R01 DK085129]
  6. Fonds de Recherche Quebec-Sante

Ask authors/readers for more resources

G protein-coupled receptors have been well described to contribute to the regulation of glucose-stimulated insulin secretion (GSIS). The short-chain fatty acid-sensing G protein-coupled receptor, free fatty acid receptor 2 (FFAR2), is expressed in pancreatic beta-cells, and in rodents, its expression is altered during insulin resistance. Thus, we explored the role of FFAR2 in regulating GSIS. First, assessing the phenotype of wild-type and Ffar2(-/-) mice in vivo, we observed no differences with regard to glucose homeostasis on normal or high-fat diet, with a marginally significant defect in insulin secretion in Ffar2(-/-) mice during hyperglycemic clamps. In ex vivo insulin secretion studies, we observed diminished GSIS from Ffar2(-/-) islets relative to wild-type islets under high-glucose conditions. Further, in the presence of acetate, the primary endogenous ligand for FFAR2, we observed FFAR2-dependent potentiation of GSIS, whereas FFAR2-specific agonists resulted in either potentiation or inhibition of GSIS, which we found to result from selective signaling through either G alpha(q/11) or G alpha(i/o), respectively. Lastly, in ex vivo insulin secretion studies of human islets, we observed that acetate and FFAR2 agonists elicited different signaling properties at human FFAR2 than at mouse FFAR2. Taken together, our studies reveal that FFAR2 signaling occurs by divergent G protein pathways that can selectively potentiate or inhibit GSIS in mouse islets. Further, we have identified important differences in the response of mouse and human FFAR2 to selective agonists, and we suggest that these differences warrant consideration in the continued investigation of FFAR2 as a novel type 2 diabetes target.

Authors

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

Reviews

Primary Rating

3.9
Not enough ratings

Secondary Ratings

Novelty
-
Significance
-
Scientific rigor
-
Rate this paper

Recommended

No Data Available
No Data Available