Normal glucose uptake in the brain and heart requires an endothelial cell-specific HIF-1α-dependent function.

TitleNormal glucose uptake in the brain and heart requires an endothelial cell-specific HIF-1α-dependent function.
Publication TypeJournal Article
Year of Publication2012
AuthorsHuang Y, Lei L, Liu D, Jovin I, Russell R, Johnson RS, Di Lorenzo A, Giordano FJ
JournalProc Natl Acad Sci U S A
Volume109
Issue43
Pagination17478-83
Date Published2012 Oct 23
ISSN1091-6490
KeywordsAnimals, Blood Glucose, Brain, Endothelial Cells, Glucose Transporter Type 1, Humans, Hypoxia-Inducible Factor 1, alpha Subunit, Male, Mice, Mice, Knockout, Myocardium
Abstract

Although intimately positioned between metabolic substrates in the bloodstream and the tissue parenchymal cells that require these substrates, a major role of the vascular endothelium in the regulation of tissue metabolism has not been widely appreciated. We hypothesized that via control of transendothelial glucose transport and contributing paracrine mechanisms the endothelium plays a major role in regulating organ and tissue glucose metabolism. We further hypothesized that the hypoxia-inducible factor -1α (HIF-1α) plays an important role in coordinating these endothelial functions. To test these hypotheses, we generated mice with endothelial cell-specific deletion of HIF-1α. Loss of HIF in the endothelium resulted in significantly increased fasting blood glucose levels, a blunted insulin response with delayed glucose clearance from the blood after i.v. loading, and significantly decreased glucose uptake into the brain and heart. Endothelial HIF-1α knockout mice also exhibited a reduced cerebrospinal fluid/blood glucose ratio, a finding consistent with reduced transendothelial glucose transport and a diagnostic criterion for the Glut1 deficiency genetic syndrome. Endothelial cells from these mice demonstrated decreased Glut1 levels and reduced glucose uptake that was reversed by forced expression of Glut1. These data strongly support an important role of the vascular endothelium in determining whole-organ glucose metabolism and indicate that HIF-1α is a critical mediator of this function.

DOI10.1073/pnas.1209281109
Alternate JournalProc Natl Acad Sci U S A
PubMed ID23047702
PubMed Central IDPMC3491491
Grant ListR01-63770 / / PHS HHS / United States
R01-64001 / / PHS HHS / United States
U24 DK59635 / DK / NIDDK NIH HHS / United States
Related Faculty: 
Annarita Di Lorenzo, Ph.D.

Pathology & Laboratory Medicine 1300 York Avenue New York, NY 10065 Phone: (212) 746-6464
Surgical Pathology: (212) 746-2700