The mechanism of insulin action on gene expression is a key question in biology, with important ramifications for the treatment of diabetes and metabolic disorders. Studies supported by this grant have established a role for the O sub-family of Forkhead transcription factors in insulin regulation of gene expression. During the past funding cycle, we have demonstrated that FoxOl is the principal insulin- dependent transcription factor in the regulation of hepatic gluconeogenesis and pancreatic (3 cell mass. We have shown that: /, phosphorylation is the main mechanism by which insulin inhibits FoxOl by promoting its nuclear exclusion;ii, FoxOl expression can single-handedlyconfer insulin regulation on the expression of Glucose-6-phosphatase, the rate-limiting enzyme in glycogenolysis;Hi,FoxOl interacts with Pgcla to stimulate gluconeogenesis;iv, FoxOl links insulin signaling to Pdxl regulation of |3cell mass. We now present preliminarydata extending the gamut of FoxO functions to regulation of cell differentiation and protection against oxidative stress, while also expanding the repertoire of FoxO target genes. We demonstrate that these functions are based on two novel modes of FoxO action: acetylation-dependent 'targeting to nuclear Pml bodies, and protein/protein interactions with the Notch effector Csl. The latter observation indicates that FoxOl functions as a coactivator, and not only as a transcription factor. It also bridges together two important signalingpathways, the PI 3-kinase and the Hesl pathways. In the next five years, we will endeavor to integrate this new information in the mechanism of FoxOl action and its role in metabolic disorders. We propose to study:
in Aim l, how phosphorylation- and acetylation-mediated mechanisms are integrated in vivo to determine the kineticsof FoxOl sub-cellularlocalization in physiologic conditions and disease states;
in Aim 2, how acetylation-dependent sub-nucleartargeting of FoxOl regulates metabolism in liver, pancreatic (3 cells and adipocytes;and in Aim 3, how the balance between the coactivator and transcription functions of Foxol is determined. The studies will be carried out with genetic loss- and gain-of-function experiments in transgenic mice and cultured cells, using methods that have been fully implemented in the Pi's laboratory. The ultimate goal of this work is to find a therapeutic approach to modifying FoxOl function. Indeed, while FoxOl is an extremely attractive biological target to treat diabetes and metabolic diseases, it is largely intractable as a drug target. Therefore, it is hoped that by identifying mechanisms of action and interacting partners, new ways to modulate its function can be found.

Agency
National Institute of Health (NIH)
Institute
National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK)
Type
Research Project (R01)
Project #
5R01DK057539-10
Application #
7750018
Study Section
Cellular Aspects of Diabetes and Obesity Study Section (CADO)
Program Officer
Silva, Corinne M
Project Start
2000-06-01
Project End
2010-12-31
Budget Start
2010-01-01
Budget End
2010-12-31
Support Year
10
Fiscal Year
2010
Total Cost
$409,937
Indirect Cost
Name
Columbia University (N.Y.)
Department
Internal Medicine/Medicine
Type
Schools of Medicine
DUNS #
621889815
City
New York
State
NY
Country
United States
Zip Code
10032
Langlet, Fanny; Tarbier, Marcel; Haeusler, Rebecca A et al. (2018) microRNA-205-5p is a modulator of insulin sensitivity that inhibits FOXO function. Mol Metab 17:49-60
Accili, Domenico (2018) Insulin Action Research and the Future of Diabetes Treatment: The 2017 Banting Medal for Scientific Achievement Lecture. Diabetes 67:1701-1709
Haeusler, Rebecca A; McGraw, Timothy E; Accili, Domenico (2018) Biochemical and cellular properties of insulin receptor signalling. Nat Rev Mol Cell Biol 19:31-44
Langlet, Fanny; Haeusler, Rebecca A; Lindén, Daniel et al. (2017) Selective Inhibition of FOXO1 Activator/Repressor Balance Modulates Hepatic Glucose Handling. Cell 171:824-835.e18
Kim-Muller, Ja Young; Kim, Young Jung R; Fan, Jason et al. (2016) FoxO1 Deacetylation Decreases Fatty Acid Oxidation in ?-Cells and Sustains Insulin Secretion in Diabetes. J Biol Chem 291:10162-72
Accili, D; Talchai, S C; Kim-Muller, J Y et al. (2016) When ?-cells fail: lessons from dedifferentiation. Diabetes Obes Metab 18 Suppl 1:117-22
Cook, Joshua R; Matsumoto, Michihiro; Banks, Alexander S et al. (2015) A mutant allele encoding DNA binding-deficient FoxO1 differentially regulates hepatic glucose and lipid metabolism. Diabetes 64:1951-65
Pajvani, Utpal B; Accili, Domenico (2015) The new biology of diabetes. Diabetologia 58:2459-68
Cook, Joshua R; Langlet, Fanny; Kido, Yoshiaki et al. (2015) Pathogenesis of selective insulin resistance in isolated hepatocytes. J Biol Chem 290:13972-80
Ren, Hongxia; Cook, Joshua R; Kon, Ning et al. (2015) Gpr17 in AgRP Neurons Regulates Feeding and Sensitivity to Insulin and Leptin. Diabetes 64:3670-9

Showing the most recent 10 out of 66 publications