Skip to main content

Lund University Publications

LUND UNIVERSITY LIBRARIES

HDL-bound sphingosine 1-phosphate acts as a biased agonist for the endothelial cell receptor S1P1 to limit vascular inflammation.

Galvani, Sylvain ; Sanson, Marie ; Blaho, Victoria A ; Swendeman, Steven L ; Conger, Heather ; Dahlbäck, Björn LU ; Kono, Mari ; Proia, Richard L ; Smith, Jonathan D and Hla, Timothy (2015) In Science Signaling 8(389). p.79-79
Abstract
The sphingosine 1-phosphate receptor 1 (S1P1) is abundant in endothelial cells, where it regulates vascular development and microvascular barrier function. In investigating the role of endothelial cell S1P1 in adult mice, we found that the endothelial S1P1 signal was enhanced in regions of the arterial vasculature experiencing inflammation. The abundance of proinflammatory adhesion proteins, such as ICAM-1, was enhanced in mice with endothelial cell-specific deletion of S1pr1 and suppressed in mice with endothelial cell-specific overexpression of S1pr1, suggesting a protective function of S1P1 in vascular disease. The chaperones ApoM(+)HDL (HDL) or albumin bind to sphingosine 1-phosphate (S1P) in the circulation; therefore, we tested the... (More)
The sphingosine 1-phosphate receptor 1 (S1P1) is abundant in endothelial cells, where it regulates vascular development and microvascular barrier function. In investigating the role of endothelial cell S1P1 in adult mice, we found that the endothelial S1P1 signal was enhanced in regions of the arterial vasculature experiencing inflammation. The abundance of proinflammatory adhesion proteins, such as ICAM-1, was enhanced in mice with endothelial cell-specific deletion of S1pr1 and suppressed in mice with endothelial cell-specific overexpression of S1pr1, suggesting a protective function of S1P1 in vascular disease. The chaperones ApoM(+)HDL (HDL) or albumin bind to sphingosine 1-phosphate (S1P) in the circulation; therefore, we tested the effects of S1P bound to each chaperone on S1P1 signaling in cultured human umbilical vein endothelial cells (HUVECs). Exposure of HUVECs to ApoM(+)HDL-S1P, but not to albumin-S1P, promoted the formation of a cell surface S1P1-β-arrestin 2 complex and attenuated the ability of the proinflammatory cytokine TNFα to activate NF-κB and increase ICAM-1 abundance. Although S1P bound to either chaperone induced MAPK activation, albumin-S1P triggered greater Gi activation and receptor endocytosis. Endothelial cell-specific deletion of S1pr1 in the hypercholesterolemic Apoe(-/-) mouse model of atherosclerosis enhanced atherosclerotic lesion formation in the descending aorta. We propose that the ability of ApoM(+)HDL to act as a biased agonist on S1P1 inhibits vascular inflammation, which may partially explain the cardiovascular protective functions of HDL. (Less)
Please use this url to cite or link to this publication:
author
; ; ; ; ; ; ; ; and
organization
publishing date
type
Contribution to journal
publication status
published
subject
in
Science Signaling
volume
8
issue
389
pages
79 - 79
publisher
American Association for the Advancement of Science (AAAS)
external identifiers
  • pmid:26268607
  • wos:000360807900001
  • scopus:84939201829
  • pmid:26268607
ISSN
1937-9145
DOI
10.1126/scisignal.aaa2581
language
English
LU publication?
yes
id
f8bfca4a-4f91-4301-b96a-b55bc485f730 (old id 7844062)
alternative location
http://www.ncbi.nlm.nih.gov/pubmed/26268607?dopt=Abstract
date added to LUP
2016-04-01 10:41:05
date last changed
2022-05-13 18:52:53
@article{f8bfca4a-4f91-4301-b96a-b55bc485f730,
  abstract     = {{The sphingosine 1-phosphate receptor 1 (S1P1) is abundant in endothelial cells, where it regulates vascular development and microvascular barrier function. In investigating the role of endothelial cell S1P1 in adult mice, we found that the endothelial S1P1 signal was enhanced in regions of the arterial vasculature experiencing inflammation. The abundance of proinflammatory adhesion proteins, such as ICAM-1, was enhanced in mice with endothelial cell-specific deletion of S1pr1 and suppressed in mice with endothelial cell-specific overexpression of S1pr1, suggesting a protective function of S1P1 in vascular disease. The chaperones ApoM(+)HDL (HDL) or albumin bind to sphingosine 1-phosphate (S1P) in the circulation; therefore, we tested the effects of S1P bound to each chaperone on S1P1 signaling in cultured human umbilical vein endothelial cells (HUVECs). Exposure of HUVECs to ApoM(+)HDL-S1P, but not to albumin-S1P, promoted the formation of a cell surface S1P1-β-arrestin 2 complex and attenuated the ability of the proinflammatory cytokine TNFα to activate NF-κB and increase ICAM-1 abundance. Although S1P bound to either chaperone induced MAPK activation, albumin-S1P triggered greater Gi activation and receptor endocytosis. Endothelial cell-specific deletion of S1pr1 in the hypercholesterolemic Apoe(-/-) mouse model of atherosclerosis enhanced atherosclerotic lesion formation in the descending aorta. We propose that the ability of ApoM(+)HDL to act as a biased agonist on S1P1 inhibits vascular inflammation, which may partially explain the cardiovascular protective functions of HDL.}},
  author       = {{Galvani, Sylvain and Sanson, Marie and Blaho, Victoria A and Swendeman, Steven L and Conger, Heather and Dahlbäck, Björn and Kono, Mari and Proia, Richard L and Smith, Jonathan D and Hla, Timothy}},
  issn         = {{1937-9145}},
  language     = {{eng}},
  number       = {{389}},
  pages        = {{79--79}},
  publisher    = {{American Association for the Advancement of Science (AAAS)}},
  series       = {{Science Signaling}},
  title        = {{HDL-bound sphingosine 1-phosphate acts as a biased agonist for the endothelial cell receptor S1P1 to limit vascular inflammation.}},
  url          = {{http://dx.doi.org/10.1126/scisignal.aaa2581}},
  doi          = {{10.1126/scisignal.aaa2581}},
  volume       = {{8}},
  year         = {{2015}},
}