The proposed study aims to determine how astrocytic leptin receptors (ObR, or LR) affect obesity regulation. Obesity, like neural injury, results in regional astrogliosis. We observed that knockout of ObR in astrocytes leads to partial resistance of the mice to diet-induced obesity, a finding opposite to the obesity found in mice with knockout of ObR in neurons. Moreover, mice with adult-onset obesity show both astrogliosis and robust upregulation of astrocytic ObR in selective nuclei of the hypothalamus. These ObR are functional, as leptin treatment activates multiple signaling pathways in primary astrocytes. We, therefore, hypothesize that the ObR (+) astrocytes provide negative regulation to facilitate obesity in two main ways: (a) reducing availability of leptin to neurons by accelerating leptin turnover;(b) modulating neuronal function by alterations of the profile of glial transmitters released. Such a role for astrocytes provides a direct mechanism linking neuroinflammatory processes and body weight control. We will test the hypotheses in three specific aims. (1) To show that reactive astrocytes facilitate the turnover of leptin and attenuate neuronal leptin signaling in the brain, Aim 1 will use an established model of reactive astrogliosis - adult-onset obesity - to determine the distribution of fluorescently conjugated leptin and pSTAT3 signaling after intracerebroventricular delivery of leptin. The results will be compared with those from astrocyte specific leptin receptor knockout mice, and after pretreatment with the astrocyte inhibitor fluorocitrate. (2) In Aim 2, we will test the hypothesis that leptin modulates the production profile of gliotransmitters, including glutamate and ATP in the acute phase and cytokines at a later time. Primary astrocytes and neurons will be used for calcium imaging, and the effects of astrocyte conditioned medium and mixed neuron-glial culture will be tested. (3) Aim 3 will identify functional consequences of astrocyte specific leptin receptor knockout on the metabolic phenotype and correlate this with serum biomarkers of neuroinflammation. Overall, the results will demonstrate an essential role of ObR(+) astrocytes in the regulation of neuronal leptin signaling. As astrocytes are crucially involved in neural injury and an integral part of the neuroimmune axis, these studies will provide tangible mechanisms by which neuroinflammation can regulate body weight.

Public Health Relevance

Leptin is a hormone mainly produced by fat tissue. Hyperleptinemia is seen in obesity and the metabolic syndrome. The incidence of obesity and its associated hyperlipidemia, cardiovascular complications, cancer, and sleep apnea have been on a steep rise nationally and globally. This study will address how astrocytes participate in delivering leptin to neurons and modulating its actions. Astrocytes are the most abundant cells in the brain, clearly involved in brain injury, but very few studies have addressed whether they are involved with leptin and obesity. We recently found that both the mRNA and protein of leptin receptors are indeed present in astrocytes. Moreover, the expression level of these leptin receptors increases in mouse models of adult-onset obesity. This suggests an important role of the astrocytic leptin system in the regulatory changes in obese subjects. A series of experiments with mice and cultured cells are designed to address how these ObR(+) astrocytes affect neuronal function and overall metabolic phenotype. Thus, their relevance lies in (a) better understanding of how astrocytes affect obesity onset and progression;(b) better understanding of cell-cell interactions in the brain;and (c) potential identification of novel therapeutic targets to better combat obesity.

Agency
National Institute of Health (NIH)
Institute
National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK)
Type
Research Project (R01)
Project #
5R01DK092245-02
Application #
8306777
Study Section
Brain Injury and Neurovascular Pathologies Study Section (BINP)
Program Officer
Hyde, James F
Project Start
2011-07-26
Project End
2015-04-30
Budget Start
2012-05-01
Budget End
2013-04-30
Support Year
2
Fiscal Year
2012
Total Cost
$327,820
Indirect Cost
$106,320
Name
Lsu Pennington Biomedical Research Center
Department
Type
Organized Research Units
DUNS #
611012324
City
Baton Rouge
State
LA
Country
United States
Zip Code
70808
He, Yi; Cornelissen-Guillaume, Germaine G; He, Junyun et al. (2016) Circadian rhythm of autophagy proteins in hippocampus is blunted by sleep fragmentation. Chronobiol Int 33:553-60
Wang, Yuping; Hsuchou, Hung; He, Yi et al. (2015) Role of Astrocytes in Leptin Signaling. J Mol Neurosci 56:829-839
Pan, Weihong (2015) From blood to brain through BBB and astrocytic signaling. Peptides 72:121-7
He, Junyun; Kastin, Abba J; Wang, Yuping et al. (2015) Sleep fragmentation has differential effects on obese and lean mice. J Mol Neurosci 55:644-52
Ouyang, Suidong; Hsuchou, Hung; Kastin, Abba J et al. (2014) Diet-induced obesity suppresses expression of many proteins at the blood-brain barrier. J Cereb Blood Flow Metab 34:43-51
Pan, Weihong; Kastin, Abba J (2014) Leptin: a biomarker for sleep disorders? Sleep Med Rev 18:283-90
Jayaram, Bhavaani; Khan, Reas S; Kastin, Abba J et al. (2013) Protective role of astrocytic leptin signaling against excitotoxicity. J Mol Neurosci 49:523-30
Hsuchou, Hung; Jayaram, Bhavaani; Kastin, Abba J et al. (2013) Endothelial cell leptin receptor mutant mice have hyperleptinemia and reduced tissue uptake. J Cell Physiol 228:1610-6
Mishra, Pramod K; Hsuchou, Hung; Ouyang, Suidong et al. (2013) Loss of astrocytic leptin signaling worsens experimental autoimmune encephalomyelitis. Brain Behav Immun 34:98-107
Wang, Yuping; He, Junyun; Kastin, Abba J et al. (2013) Hypersomnolence and reduced activity in pan-leptin receptor knockout mice. J Mol Neurosci 51:1038-45

Showing the most recent 10 out of 21 publications