Endothelial S1P Signaling Counteracts Infarct Expansion in Ischemic Stroke.

TitleEndothelial S1P Signaling Counteracts Infarct Expansion in Ischemic Stroke.
Publication TypeJournal Article
Year of Publication2021
AuthorsNitzsche A, Poittevin M, Benarab A, Bonnin P, Faraco G, Uchida H, Favre J, Garcia-Bonilla L, Garcia MCL, Léger P-L, Thérond P, Mathivet T, Autret G, Baudrie V, Couty L, Kono M, Chevallier A, Niazi H, Tharaux P-L, Chun J, Schwab SR, Eichmann A, Tavitian B, Proia RL, Charriaut-Marlangue C, Sanchez T, Kubis N, Henrion D, Iadecola C, Hla T, Camerer E
JournalCirc Res
Volume128
Issue3
Pagination363-382
Date Published2021 02 05
ISSN1524-4571
KeywordsAnimals, Blood-Brain Barrier, Cerebral Arteries, Cerebrovascular Circulation, Disease Models, Animal, Endothelial Cells, Female, Infarction, Middle Cerebral Artery, Ischemic Attack, Transient, Ischemic Stroke, Lysophospholipids, Male, Mice, 129 Strain, Mice, Inbred C57BL, Mice, Knockout, Microcirculation, Neuroprotective Agents, Signal Transduction, Sphingosine, Sphingosine-1-Phosphate Receptors, Vascular Patency
Abstract

RATIONALE: Cerebrovascular function is critical for brain health, and endogenous vascular protective pathways may provide therapeutic targets for neurological disorders. S1P (Sphingosine 1-phosphate) signaling coordinates vascular functions in other organs, and S1P (S1P receptor-1) modulators including fingolimod show promise for the treatment of ischemic and hemorrhagic stroke. However, S1P also coordinates lymphocyte trafficking, and lymphocytes are currently viewed as the principal therapeutic target for S1P modulation in stroke.

OBJECTIVE: To address roles and mechanisms of engagement of endothelial cell S1P in the naive and ischemic brain and its potential as a target for cerebrovascular therapy.

METHODS AND RESULTS: Using spatial modulation of S1P provision and signaling, we demonstrate a critical vascular protective role for endothelial S1P in the mouse brain. With an S1P signaling reporter, we reveal that abluminal polarization shields S1P from circulating endogenous and synthetic ligands after maturation of the blood-neural barrier, restricting homeostatic signaling to a subset of arteriolar endothelial cells. S1P signaling sustains hallmark endothelial functions in the naive brain and expands during ischemia by engagement of cell-autonomous S1P provision. Disrupting this pathway by endothelial cell-selective deficiency in S1P production, export, or the S1P receptor substantially exacerbates brain injury in permanent and transient models of ischemic stroke. By contrast, profound lymphopenia induced by loss of lymphocyte S1P provides modest protection only in the context of reperfusion. In the ischemic brain, endothelial cell S1P supports blood-brain barrier function, microvascular patency, and the rerouting of blood to hypoperfused brain tissue through collateral anastomoses. Boosting these functions by supplemental pharmacological engagement of the endothelial receptor pool with a blood-brain barrier penetrating S1P-selective agonist can further reduce cortical infarct expansion in a therapeutically relevant time frame and independent of reperfusion.

CONCLUSIONS: This study provides genetic evidence to support a pivotal role for the endothelium in maintaining perfusion and microvascular patency in the ischemic penumbra that is coordinated by S1P signaling and can be harnessed for neuroprotection with blood-brain barrier-penetrating S1P agonists.

DOI10.1161/CIRCRESAHA.120.316711
Alternate JournalCirc Res
PubMed ID33301355
PubMed Central IDPMC7874503
Grant ListR01 NS034179 / NS / NINDS NIH HHS / United States
R01 NS114561 / NS / NINDS NIH HHS / United States
ZIA DK056014 / ImNIH / Intramural NIH HHS / United States
Related Faculty: 
Teresa Sanchez, Ph.D.

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