Obesity is clearly one of the most visible public health problems in the US. Obesity now affects more than 30 % of the US adult population, increasing the risks for serious chronic diseases and reducing the life expectancy. This highlights urgent need to better understand the etiology of obesity and develop more effective therapies against obesity. Leptin is a key adipocyte-derived hormone that potently suppresses food intake, reduces body weight, and increases energy expenditure. Thus, leptin was once thought to be a magic bullet for the treatment of obesity. However, leptin does not work in obese people because of the development of leptin resistance. This observation creates one of the fundamental questions to be addressed in the field: what are the mechanisms of neuronal leptin resistance, a hallmark of human obesity. To address this question, we have been using an organotypic brain slice model as an in vitro tool to investigate the molecular mechanisms underlying cellular leptin resistance in hypothalamic neurons, a primary site of leptin action. Using this tool, we initially found that the cAMP-related pathway potently induces leptin resistance through Epac-Rap1 signaling. Epac is an exchange factor for GTP/GDP for the small G protein Rap1. Furthermore, we have searched for an extracellular upstream factor(s) that induces both the activation of Epac-Rap1 signaling and leptin resistance. To this end, we have been conducting a candidate-ligand approach based on the fact that Epac-Rap1 signaling can be activated by a variety of G protein-coupled receptors (GPCRs) that produce cAMP. We have been systematically screening the ligands of known GPCRs that couple to cAMP signaling. During our initial screening, we have identified the gut hormone glucose-dependent insulinotropic polypeptide (GIP) as a promising candidate. Based on these previous observations and our preliminary data, we hypothesize that the gut-derived GIP acts as a previously unrecognized circulating signal that drives neuronal leptin resistance via directly activating Epac-Rap1 signaling during obesity. In our specific aims, in Aim1, we will use a Cre- dependent conditional Rap1 knockout mouse to determine the physiological relevance of Rap1 expressed by leptin responsive neurons in diet-induced leptin resistance and obesity.
In Aim2, we will determine if GIP receptor in leptin responsive neurons is required for diet-induced leptin resistance and obesity by using GIP receptor deficient mice.
In Aim3, through a combination of pharmacological and genetic studies in rodents, we will also explore potential intervention strategies targeting GIP receptor and Epac, and using the same approach, we will determine the molecular mechanisms mediating the effect of brain GIP-Epac-Rap1 signaling on modulating leptin sensitivity. The results of these studies are potentially paradigm-shifting, and should provide a framework for a better understanding of central leptin resistance.

Public Health Relevance

Reduction in body weight has a beneficial impact on a number of chronic diseases including type II diabetes. Leptin would be an attractive therapeutic target to combat obesity when leptin responsiveness in obesity can be restored. The successful completion of this project will advance our understanding of leptin resistance and will provide new strategies to restore leptin responsiveness and thus counteract obesity.

Agency
National Institute of Health (NIH)
Institute
National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK)
Type
Research Project (R01)
Project #
1R01DK104901-01A1
Application #
9026967
Study Section
Integrative Physiology of Obesity and Diabetes Study Section (IPOD)
Program Officer
Hyde, James F
Project Start
2015-09-24
Project End
2020-07-31
Budget Start
2015-09-24
Budget End
2016-07-31
Support Year
1
Fiscal Year
2015
Total Cost
$315,000
Indirect Cost
$90,000
Name
Baylor College of Medicine
Department
Pediatrics
Type
Schools of Medicine
DUNS #
051113330
City
Houston
State
TX
Country
United States
Zip Code
77030
Kaneko, Kentaro; Xu, Pingwen; Cordonier, Elizabeth L et al. (2016) Neuronal Rap1 Regulates Energy Balance, Glucose Homeostasis, and Leptin Actions. Cell Rep 16:3003-3015
Saito, Kenji; He, Yanlin; Yang, Yongjie et al. (2016) PI3K in the ventromedial hypothalamic nucleus mediates estrogenic actions on energy expenditure in female mice. Sci Rep 6:23459
Zhu, Liangru; Xu, Pingwen; Cao, Xuehong et al. (2015) The ER?-PI3K Cascade in Proopiomelanocortin Progenitor Neurons Regulates Feeding and Glucose Balance in Female Mice. Endocrinology 156:4474-91