Obesity is currently a worldwide epidemic, therefore preventing and reversing obesity is of utmost importance for the general health of the population. Body weight regulation is maintained through a balance of energy intake versus energy expenditure, and targeting appetite or metabolic pathways via obesity therapeutics is a current goal in obesity research. The brain is the center for control and coordination of body weight. Hypothalamic melanocortin system activation reduces appetite, and also sends signals to the nucleus of the solitary tract (NTS) which activates sympathetic nervous system (SNS) innervation of brown adipose tissue (BAT), leading to increased thermogenesis (energy expenditure).
The aim of this proposal is to further investigate a family of molecules recently found to be involved in metabolism. The bone morphogenetic proteins (BMPs) are growth factors, and recent studies have demonstrated roles for the BMPs in metabolism and satiety in lower organisms. Our laboratory has also shown that BMP7 in mice is able to stimulate brown adipocyte differentiation and to induce BAT thermogenesis. Additionally, we have shown that BMP7 and its receptors are expressed in the hypothalamus and BMP7 delivery (either i.c.v. or systemically via adenovirus) produces an anorexigenic effect, at least in part through the hypothalamic mTOR pathway and activation of the melanocortin system. However, much is still unknown about the metabolic and physiological roles of BMP signaling in energy balance. Therefore, the overarching goal of this project is to identify how BMP7 impacts whole body energy balance by affecting appetite pathways and BAT thermogenesis. Thus far, we have found that mice with BMP7 haploinsufficiency (BMP7-/+) on a high fat diet (HFD) are more obese and hyperphagic than littermate controls, with decreased energy expenditure (oxygen consumption/VO2) levels. Therefore, our first objective is to determine the physiological mechanism for BMP7's effects on energy balance, with the hypothesis that BMP7 is regulating the hypothalamic-NTS melanocortin system which regulates both central appetite pathways as well as energy expenditure pathways (via sympathetic innervation of BAT). We have also shown that mice with POMC-neuron (ie: in hypothalamus and NTS/brainstem) deletion of BMPR1a (a BMP7 receptor) exhibit hyperphagia as well as increased energy expenditure and BAT thermogenesis. Therefore our second objective is to determine whether the mechanism for POMC-neuron BMPR1a action is through hypothalamic melanocortin pathway inhibition to induce hyperphagia, and through the NTS-SNS pathways to increase thermogenesis in BAT. These studies will utilize novel mouse models for metabolic studies, including an important model linking central BMP signaling disruption with peripheral energy expenditure effects, and will investigate a novel class of appetite factors: the BMP growth factors. Therefore, the research outlined in this proposal could provide important new findings for the field of obesity research, and potentially lead to development of new obesity therapeutics.

Public Health Relevance

Obesity is currently epidemic worldwide, therefore current biomedical science seeks to develop obesity therapeutics and to understand the pathophysiology of obesity in order to implement prevention strategies. This project utilizes two novel mouse models to investigate the role of the BMPs, a family of growth factors, in both nervous system and brown adipose tissue pathways regulating body weight homeostasis.

Agency
National Institute of Health (NIH)
Institute
National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK)
Type
Postdoctoral Individual National Research Service Award (F32)
Project #
1F32DK091996-01A1
Application #
8202892
Study Section
Special Emphasis Panel (ZDK1-GRB-9 (M1))
Program Officer
Castle, Arthur
Project Start
2011-06-07
Project End
2013-06-06
Budget Start
2011-06-07
Budget End
2012-06-06
Support Year
1
Fiscal Year
2011
Total Cost
$53,042
Indirect Cost
Name
Joslin Diabetes Center
Department
Type
DUNS #
071723084
City
Boston
State
MA
Country
United States
Zip Code
02215
Townsend, Kristy L; Madden, Christopher J; Blaszkiewicz, Magdalena et al. (2017) Reestablishment of Energy Balance in a Male Mouse Model With POMC Neuron Deletion of BMPR1A. Endocrinology 158:4233-4245
Townsend, K L; Tseng, Y-H (2015) Of mice and men: novel insights regarding constitutive and recruitable brown adipocytes. Int J Obes Suppl 5:S15-20
Stanford, Kristin I; Middelbeek, Roeland J W; Townsend, Kristy L et al. (2015) A novel role for subcutaneous adipose tissue in exercise-induced improvements in glucose homeostasis. Diabetes 64:2002-14
Zhang, Hongbin; Guan, Meiping; Townsend, Kristy L et al. (2015) MicroRNA-455 regulates brown adipogenesis via a novel HIF1an-AMPK-PGC1? signaling network. EMBO Rep 16:1378-93
Townsend, Kristy L; Tseng, Yu-Hua (2014) Brown fat fuel utilization and thermogenesis. Trends Endocrinol Metab 25:168-77
Schulz, Tim J; Huang, Ping; Huang, Tian Lian et al. (2013) Brown-fat paucity due to impaired BMP signalling induces compensatory browning of white fat. Nature 495:379-83
Townsend, Kristy L; An, Ding; Lynes, Matthew D et al. (2013) Increased mitochondrial activity in BMP7-treated brown adipocytes, due to increased CPT1- and CD36-mediated fatty acid uptake. Antioxid Redox Signal 19:243-57
Townsend, Kristy L; Suzuki, Ryo; Huang, Tian Lian et al. (2012) Bone morphogenetic protein 7 (BMP7) reverses obesity and regulates appetite through a central mTOR pathway. FASEB J 26:2187-96