In the third grant cycle of leptin transport across the BBB , we will focus on the role of leptin receptor (ObR)- positive astrocytes in relaying leptin from blood to the CNS by crossing the blood-brain barrier (BBB). We hypothesize that astrocytes not only regulate leptin transport as vital components of the BBB, but also modulate neuronal leptin signaling by enabling a more rapid onset and faster termination of leptin action in neurons. The cellular studies will use a co-culture model of primary cerebral microvascular endothelial cells (PBMEC) and astrocytes. We will determine the effects of astrocytes on the permeability and integrity of fluorescently or radioactively labeled leptin and the permeability markers dextran and albumin across the tight monolayer of PBMEC. The mouse studies will test the effects of obesity-induced increase of astrocytic ObR expression, inhibition of astrocyte metabolic activity, and specific deletion of ObR from astrocytes on leptin distribution and neuronal activation in response to leptin.
In Aim 1, we will test the hypothesis that astrocytes thwart leptin transport across the PBMEC monolayer by slowing down permeation kinetics. The transcellular rather than paracellular pathways will be shown by morphological examination of the location of fluorescently labeled leptin, as well as the permeation kinetic studies to test the specificity of transport. The effect the level of astrocytic ObR expression will be tested. The transport of leptin will be compared with the distribution of co-administered paracellular permeability markers and immunocytochemistry of tight junction proteins.
In Aim 2, we will test the hypothesis that astrocytes and astrocytic ObR participate in the degradation of leptin within the CNS, modulate the half-life of leptin in neurons, and influence neuronal leptin signaling by generation of soluble glial transmitters.
Aim 3 will focus on regulatory changes in adult mice with diet-induced obesity or the Avy mutation, both of which have been shown to have regional specific increases of astrocytic ObR. By use of glial metabolic inhibitors and newly generated astrocyte-specific ObR knockout mice, we will show that these ObR(+) astrocytes play an essential role in the regulation of neuronal leptin signaling. The results will provide the first evidence of the functions of ObR(+) astrocytes in linking BBB transport to the CNS response to leptin. An understanding of the consequence of astrogliosis and upregulation of astrocytic ObR in obesity should enable the targeting of astrocytes to counteract the neuroendocrine dysregulation in obese subjects.

Public Health Relevance

Leptin is a hormone mainly produced by fat tissue. Hyperleptinemia is seen in the metabolic syndrome. Obesity and its associated hyperlipidemia, cardiovascular complications, cancer, and sleep apnea have a rapidly increasing prevalence in the US and many other parts of the world. This study will mainly focus on how astrocytes participate in getting leptin from blood to brain and allowing it to reach neurons, the most commonly considered effector cells. Astrocytes are the most abundant cells in the brain, but very few studies have addressed whether they have anything to do with leptin. 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. It is possible that it is neuron-glial interactions, rather than direct activation of neurons, that play important mediatory roles for blood-borne leptin. Thus, the 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
High Priority, Short Term Project Award (R56)
Project #
2R56DK054880-11
Application #
8074152
Study Section
Brain Injury and Neurovascular Pathologies Study Section (BINP)
Program Officer
Hyde, James F
Project Start
2010-07-05
Project End
2011-06-30
Budget Start
2010-07-05
Budget End
2011-06-30
Support Year
11
Fiscal Year
2010
Total Cost
$222,000
Indirect Cost
Name
Lsu Pennington Biomedical Research Center
Department
Type
Organized Research Units
DUNS #
611012324
City
Baton Rouge
State
LA
Country
United States
Zip Code
70808
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
He, Junyun; Hsuchou, Hung; He, Yi et al. (2014) Sleep restriction impairs blood-brain barrier function. J Neurosci 34:14697-706
Pan, Weihong; Kastin, Abba J (2014) Leptin: a biomarker for sleep disorders? Sleep Med Rev 18:283-90
Pan, Weihong; Kastin, Abba J (2014) Can sleep apnea cause Alzheimer's disease? Neurosci Biobehav Rev 47:656-69
He, Junyun; Wang, Yuping; Kastin, Abba J et al. (2014) Increased sleep fragmentation in experimental autoimmune encephalomyelitis. Brain Behav Immun 38:53-8
Ouyang, Suidong; Hsuchou, Hung; Kastin, Abba J et al. (2014) Leukocyte infiltration into spinal cord of EAE mice is attenuated by removal of endothelial leptin signaling. Brain Behav Immun 40:61-73
Hsuchou, Hung; Wang, Yuping; Cornelissen-Guillaume, Germaine G et al. (2013) Diminished leptin signaling can alter circadian rhythm of metabolic activity and feeding. J Appl Physiol (1985) 115:995-1003

Showing the most recent 10 out of 46 publications