Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2021 Sep 1.
Published in final edited form as: Biochim Biophys Acta Mol Cell Biol Lipids. 2020 Jun 22;1865(9):158760. doi: 10.1016/j.bbalip.2020.158760

Post-translational modifications of S1PR1 and endothelial barrier regulation

Mumtaz Anwar 1, Dolly Mehta 1,*
PMCID: PMC7409382  NIHMSID: NIHMS1606276  PMID: 32585303

Abstract

Sphingosine-1-phosphate receptor-1 (S1PR1), a G-protein coupled receptor that is expressed in endothelium and activated upon ligation by the bioactive lipid sphingosine-1-phosphate (S1P), is an important vascular-barrier protective mechanism at the level of adherens junctions (AJ). Loss of endothelial barrier function is a central factor in the pathogenesis of various inflammatory conditions characterized by protein-rich lung edema formation, such as acute respiratory distress syndrome (ARDS). While several S1PR1 agonists are available, the challenge of arresting the progression of protein-rich edema formation remains to be met. In this review, we discuss the role of S1PRs, especially S1PR1, in regulating endothelial barrier function. We review recent findings showing that replenishment of the pool of cell-surface S1PR1 may be crucial to the effectiveness of S1P in repairing the endothelial barrier. In this context, we discuss the S1P generating machinery and mechanisms that regulate S1PR1 at the cell surface and their impact on endothelial barrier function.

Introduction

Sphingosine 1 phosphate receptor 1 (S1PR1), upon ligating S1P, regulates a variety of endothelial functions including barrier function, vasculogenesis, vascular tone, inflammation, lymphocyte trafficking, chemotaxis and immunity [14]. S1PR1 belongs to the family of seven-transmembrane-domain G-protein coupled receptors (GPCRs) [5], the largest family of membrane proteins in the human genome to transmit diverse signals from extracellular GPCR ligands [6]. So far, 5 S1PRs (S1PR1-S1PR5) (formerly termed endothelial differentiation gene (Edg) receptors) are characterized[7]. Additionally, three putative but less well-characterized S1P-GPCRs have been identified (GPR3, GPR6, and GPR12) [2, 8]. S1PRs share 40% similarity to lysophosphatidic acid (LPA)-activated receptors [9]. S1PRs 1–3 are highly expressed and distributed widely in immune, cardiovascular and nervous systems [10]. In contrast, S1PR4 and S1PR5 expression is variable within tissues and are shown to be expressed in lymphatic and nervous system, respectively, but at lower levels than S1PRs 1–3. S1PR1 predominantly couples with the Gi/o alpha subunit of heterotrimeric G-proteins, whereas S1PR2 and S1PR3 couple with Gi/o, Gq and G12/13, and S1PR4 and S1PR5 with Gi/o and G12/13 [11]. Thus, differential patterns of S1PR expression within tissues along with the diversity of G-proteins to which they couple dictate their varied organ functions. S1P concentrations in the circulation approach ~1 μM, while lymph contains ~100 nM S1P[12]. However, S1PRs including S1PR1 are activated by S1P concentrations within sub-nanomolar range [13]. Interestingly, as we will discuss, mechanisms exist to downregulate the receptor response to S1P by modulating the expression of S1PR1 on the endothelial cell-surface. Herein, we review the role of the S1P-S1PRs pathway with specific focus on recent advances describing the role of post-translational mechanisms in regulating S1PR1 activity and how post-translationally modified S1PR1 alters endothelial barrier function.

Endothelial Barrier

The vascular endothelium, the innermost lining of all blood vessels, allows size-dependent transport of fluids, ions, macromolecules and leukocytes across the vessel wall by dynamically regulating the opening of intercellular junctions or through vesicles such as caveolae[3, 14]. Endothelial barrier function is regulated via the transcellular and the paracellular pathways. The transcellular pathway, also known as transcytosis, is a constitutive process that transports macromolecules larger than 3 nM through caveolae and traffics albumin and other nutrients across the endothelial wall, crucial for cellular and tissue functions, in an energy dependent manner [15, 16]. The paracellular pathway is regulated by intercellular endothelial junctions known as tight junctions, adherens junctions and gap junctions [3, 14]. In endothelial cells (EC), adherens junctions (AJ) formed by vascular endothelial (VE) cadherins along with their catenins play a predominant role in creation of the endothelial barrier while allowing passive transport of solutes/ions smaller than 3 nM in diameter. Pro-inflammatory mediators secreted during tissue injury such as thrombin, histamine, platelet activating factor and vascular endothelial growth factor (VEGF) as well as cytokines-chemokines weaken AJ, leading to an increase in endothelial permeability. Persistent opening of AJ leads to the accumulation of protein-rich edema fluid in the interstitial space, a hallmark of vascular permeability disorders including atherosclerosis and acute respiratory distress syndrome (ARDS) [3, 14, 17, 18]. S1P in general enhances barrier function and also rescues endothelial barrier function after injury by inflammatory mediators [17, 19], however, it can also disrupt barrier function depending on which receptors are being ligated and activated.

Overview of S1P Receptors in regulating endothelial barrier function

A detailed view of the S1P receptors along with their agonists and antagonists have been provided in Table 1. S1PR1 is required for development of blood vessel, since global or endothelial specific deletion of S1PR1 in mice induced embryonic lethality due to severe embryonic hemorrhage [20, 21]. In addition, EC-S1PR1 prevented aberrant angiogenesis in the postnatal embryos by preventing endothelial barrier disruption by vascular endothelial growth factor receptor 2 (VEGFR2) [18, 22]. Studies showed that in the absence of EC-S1PR1, VEGFR2-mediated tyrosine phosphorylation of VE-cadherin at Y638 led to disruption of vascular barrier function enabling VEGFR2 to induce aberrant angiogenesis [22, 23]. However, EC-S1PR1 supported VEGFR2-mediated angiogenic signaling during tumor growth [24, 25]. Balaji et al showed using a xenograft model in a S1PR1 GFP-reporter mice that cancer cells induced S1PR1-Gi signaling in EC which cooperated with VEGF-VEGFR2 signaling. These authors showed that EC-S1PR1 functioned by augmenting the activity of the tyrosine kinase c-Abl1. cAbl1 phosphorylated VEGFR2 at tyrosine-951, which prolonged VEGFR2 retention on the plasmalemma to signal sustained EC migration and vascularization and thereby tumor growth. Thus, mice lacking S1PR1 in EC showed reduced tumor vascularization and stunted growth. It seems therefore that EC-S1PR1 regulates VEGFR2 signaling in a cell-context dependent manner. While S1PR1 restricts VEGFR2 angiogenic activity during development it facilitates pathological angiogenesis by co-operating with VEGFR2 [18, 22, 23, 25, 26].

Table 1.

shows S1PRs tissue distribution and documented agonists and antagonists

Receptors Distribution Agonists Anatagonists
S1PR1 Most tissues, cerebellum, Plasma membrane, vesicles, cytoplasmic, nucleus/perinuclear region [27, 28] FTY720-P [29], AUY954 [30], KRP-203 [31], RP-002 [20], SEW2871CYM5442 Arylpropionic acids RP-001 [32] (R)-W146a or [ML056] [33, 34], VPC-23019 and VPC44116 [35]
S1PR2 Most tissues, Plasma membrane, cytoplasm [36] ML031[37] JTE-013 [38, 39]
S1PR3 Highest in lung, spleen, intestine and kidney, Plasma membrane [40] FTY720-P KRP-203 ML249 [41] VPC-23019 TY-52156 [42]
S1PR4 Lymphoid tissues and blood cells [43] FTY720-P KRP-203 ML178 [44], ML248 [45] ML131 [13, 45]
S1PR5 Brain, skin and NK cells expressing mostly [43, 46] FTY720-P KRP-203 [41]

Several endothelial cell types, including pulmonary endothelial cells, typically express the S1P1, S1P2, and S1P3 receptors[10]. However, siRNA mediated depletion of S1PR1 or EC-specific loss of S1PR1 constitutively disrupted EC-barrier function [47]. Conversely, activation of S1PR1 using S1P or a S1PR1 agonist rescued endothelial barrier function following injury induced by thrombin, LPS or Pseudomonas aeriginosa [47, 48]. These findings suggest that S1PR1 is primarily required to maintain endothelial barrier function constitutively and after inflammatory insults. Consistent with this idea, EC-S1PR1 activity limited leukocyte transmigration and prevented inflammatory injury [49, 50]. However, it seems that timing of administration of S1PR1 activity may be the factor in dictating the anti-vascular injury potential of activated EC-S1PR1 since a recent study showed that SEW2871, a S1PR1 agonist, failed to attenuate inflammation-induced endothelial barrier breakdown in a sepsis model [51].

Upon ligating S1P, S1PR1 strengthens endothelial barrier by stimulating the G-protein Gαi, which in turn induces intracellular Ca2+ transients, activation of the small GTPases Rac1 and Cdc42 as well as phosphatidylinositol 3-kinase (PI3K), and ERK[5255] (Fig. 1). Activated Rac1 leads to cortical actin assembly and reannealing of endothelial AJ in part by inhibiting VE-cadherin disassembly from junctions [53, 56, 57]. Activated Rac1 can also promote AJ assembly through suppressing RhoA activity at the level of junctions [56, 58]. Evidence also indicates that oxidized lipids can employ S1PR1 pathway to enhance barrier function. For example, Singleton et al showed that transactivation of S1PR1 by OxPAPC (oxidized 1-palmitoyl-2-arachidonoyl-sn-glycero-3-phosphocholine) induced rapid recruitment of S1PR1 to caveolin-enriched microdomains, which enhanced barrier function[59].

Figure 1: Role of S1PRs signaling regulating endothelial barrier function.

Figure 1:

Upon activation, sphingosine kinase (SPHK1) synthesizes S1P from sphingosine (SPH). S1P is transported through the transporter, SPNS2, and ligates S1PR1, S1PR2 or S1PR3. S1PR1 stimulates Gi which in turn induces a barrier protective signaling including Ca2+ mobilization and PI3-kinase activity leading to activation of small GTPase Rac1. Rac1 anneals adherens junctions enhancing barrier function. S1P also ligates S1PR2 and S1PR3, which in contrast to S1PR1, induces G12/13 activity. Activated G12/G13 stimulates Rho and ROCK signaling which destabilize adherens junctions in part by counteracting Rac1 activity through Rac1GTPase (Rac1GAP) hence disrupting barrier function. S1PR3 interacts with Gq leading to induction of Ca2+ signaling pathway that in addition to inducing Rho/ROCK pathway activates endothelial nitric oxide synthase (e-NOS). eNOS leads to NO generation which disrupts adherens junctions by nitosylating p190RhoGAP, a GTPase that blocks RhoA activity.

Studies also showed that S1P enhanced transendothelial electrical resistance, a measure of endothelial barrier function, more in pulmonary microvascular cells than in pulmonary arterial endothelial cells, indicating that the S1PR1 expression profile may differ among EC from various vascular beds [60, 61]. Recently, Kono et al, developed a mouse model based on the “Tango system”[62], in which S1PR1 activity can be tracked by following generation of nuclear GFP upon activation of β-arrestin by S1PR1 [26, 63]. Using this murine model, Engelbrecht E et al performed single cell RNAseq of mouse aorta which revealed 8-transcriptomically distinct EC subtypes [64]. One of these arterial EC subset at vascular branch point seem to show ligand-independent S1PR1/β-arrestin coupling while the other EC subtype at non-branch point showed an inflammatory gene expression signature. Akhter et al also used this model and showed that S1PR1 activation occurs dynamically in resident EC during the repair phase of injury post-LPS challenge. Interestingly, activated S1PR1 directed resident EC to acquire a reparative lineage capable of forming vasculature de novo and repairing the damaged endothelial barrier (Akhter et al un-published data). Further, studies on subsegmental expression profiling of S1PR1 in vivo will define how S1PR1 contributes to the differences in vascular permeability in various vascular beds.

S1PR2 was one of the first S1PR to be identified as a S1P receptor and was previously also known as AGR16, H218, or lpB2 [65]. Initially, S1PR2 was shown to be important for the development of heart in zebrafish and thereby their survival by promoting migration of myocardial precursor cells[42, 66]. However, later studies showed that disruption of the S1PR2 had no effect on mouse embryonic development [67] but did decrease vascular tone in response to vasoconstrictor agents [68], indicating that S1PR2 may regulate cardiovascular function [69]. Additionally, deletion of S1PR2 interfered with formation of the vasculature in the inner ear [70]. In contrast to S1PR1, S1PR2 was shown to suppress pathological angiogenesis. Studies showed that targeted disruption of the S1PR2 in mice led to a high incidence of diffuse, large B-cell lymphoma (DLBCL) of germinal center (GC) origin [71, 72]. Moreover, S1PR2 was found to be mutated in 26% of human DLBCL [71, 72]. While mechanism remains to parse out evidence indicates that S1PR2 may prevent DLBCL by tightly confining B cells in lymphoid organs via suppressing Akt activity[72]. Activated S1PR2 signals by stimulating Gαi, Gαq and Gα12/13[73, 74]. Thus, S1P ligation of S1PR2 induces downstream signaling, some of which opposes S1PR1-mediated pathways such as induction of vascular permeability through activation of the phosphatase and tensin homologue (PTEN), RhoGTPase and its downstream effector, RHO-associated protein kinase (ROCK) [37, 75]. Consistent with these findings, S. pneumoniae infection in mice increased S1PR2 expression, which in turn induced inflammatory lung injury by pathway involving the Rho-ROCK pathway [76]. Similarly, inhibition of S1PR1 activity using VPC 23019 markedly enhanced venular leakage in response to histamine while inhibition of S1PR2 signaling by JTE-013, a specific antagonist of S1PR2, counteracted the histamine-induced microvascular permeability response [77]. Also, silencing of S1PR2 significantly attenuated EC barrier enhancement by both S1P and hepatocyte growth factor (HGF) [78]. These studies indicate that a balance between S1PR1 and S1PR2 signaling may be required to regulate vascular homeostasis.

S1PR3, earlier known as lpB3, also couples to Gαq, Gαi, and Gα12/13[73]. S1PR3 was shown to play opposite roles in arterial injury. Thus, neointimal hyperplasia following denudation of iliac-femoral arteries was suppressed in mice lacking S1PR3 [79]. However, in another study S1PR3-null mice showed increased lesion size in carotid arteries following ligation injury [80]. These discrepancies could be due to higher expression of S1PR3 expression in iliac-femoral arteries versus carotid arteries [80]. It is also possible that EC expressing S1PR3 may have interfered with the injury response in carotid arteries. Furthermore, S1PR3 expression was recently shown to be a biomarker of sepsis and mortality in intensive care patients [81]. Thus, these authors showed that S1PR3 was shed from ECs following LPS-induced lung injury. Hence, S1PR3 containing microparticles disrupted EC-barrier function. Similarly, Niessen et al have shown that mice lacking S1PR3 or treatment of mice with S1PR3 specific antagonists protected them from lethal sepsis induced by LPS [82].

Much less is known about the role of S1PR4–5 in regulating endothelial barrier function. S1PR4, also known as lpC1 [83], couples to Gαi and Gα12/13 and appears to be crucial for megakaryocyte differentiation, platelet formation and activation [84]. S1PR4 activation by VPC23153 induced vasoconstriction in rat vessels [85]. S1PR4 also induced the release of IL-10 and specifically inhibited the proliferation of T cells and release of effector cytokines [86, 87], indicating T-cell immunomodulatory effect of fingolimod may in part be mediated through both S1PR1 and S1PR4 [88, 89].

S1PR5, earlier known as lpB4 or Nrg-1, couples to both Gαi and Gα12/13. Evidence indicates that S1PR5 is expressed in brain endothelial cells and suppresses the migration of oligodendrocyte precursor cells through a Rho kinase-dependent pathway, indicating that it may have a role in cellular communication during brain development and in mediating immune quiescence and integrity of blood-brain-barrier [9092]. Consistent with these findings, A-971432, the only selective agonist of S1PR5, improved blood-brain-barrier integrity in mice and reversed age-related cognitive decline [93].

Role of S1P metabolism in regulating S1PRs function

S1P is produced by almost all cells intracellularly as an intermediate of membrane sphingolipid metabolism [94]. S1P is generated by phosphorylation of sphingosine (SPH) [95]. SPH is generated by stepwise catalysis of sphingomyelin into ceramide, the later acting as a precursor for synthesis of SPH [96]. Exported S1P can activate all S1PRs in a paracrine-autocrine manner activates S1PRs [97]. Intracellular S1P can associate with specific intracellular target proteins, such as human telomerase reverse transcriptase (hTERT), HDACs or stimulator of interferon gene (STING), to induce various cellular responses [98100]. The half-life of S1P in the circulation is very low (~15 minutes), indicating that vascularized tissue must have high S1P synthetic activity to maintain plasma S1P. In this context, the EC and erythrocytes are the major source of S1P in plasma under physiologic conditions whereas platelets become an important source of S1P under pathological conditions [101, 102].

The conversion of sphingosine into S1P is mediated by one of the two sphingosine kinases (SPHK1 or SPHK2) [95, 103]. SPHK1 and SPHK2 are well-characterized biochemically and are synchronized in a spatial and temporal fashion by various modifications at the post-translational level and also by interaction with a particular set of proteins and lipids [104, 105]. In contrast to SPHK1, SPHK2 contains ~240 more amino acids [106]. Deletion of either SPHK1 or SPHK2 decreased plasma S1P by 50% without producing any defects in vasculature development, indicating that there is a high degree of functional redundancy between the two enzymes in generating S1P and thereby in regulation of S1PR activity [107, 108]. However, deletion of both SPHK1 and SPHK2 induced embryonic death of all homozygous pups by E13.5 due to poor development of the aorta, specifically incomplete pericyte coverage of blood vessels, severe hemorrhage of the brain (primarily mesenchymal) and to some extent spinal cord, mandible and pericardium [39, 109]. In unstimulated cells, SPHK1 is mainly present in the cytosol and can be exported through vesicles [110112]. Upon agonist stimulation, ERK phosphorylates SPHK1 at serine 225 [113, 114]. Phosphorylated SPHK1 translocates to the plasma membrane or exported extracellularly to increase S1P generation [111114]. Intriguingly, membrane translocation of SPHK1 was shown to be coupled with endocytic membrane trafficking [115], indicating that it has a role beyond paracrine S1P signaling [116]. In EC, SPHK1 regulation of endothelial barrier function is known to occur through paracrine signaling, since thrombin and LPS increased SPHK1 activity but decreased the intracellular S1P concentration that normally precedes the onset of barrier recovery [117].

SPHK2 is localized within membranous compartments such as endoplasmic reticulum (ER), mitochondria and nucleus [118, 119]. Thus, in contrast to SPHK1, SPHK2 induction of S1P within intracellular organs serves to regulate cellular functions directly through binding the effector proteins independent of S1PRs activation. For example, studies showed that SPHK2-induced S1P locally interacted with HDAC1 and HDAC2 in the nucleus to enhance pro-inflammatory gene transcription in epithelial cells inducing thereby lung injury [37, 120122]. SPHK2 induced S1P also bound to hTERT allosterically promoting telomerase stability and hence telomere maintenance, cell proliferation, and tumor growth [98, 99] [37, 120]. However, a recent study showed that in macrophages SPHK2 was required for inhibiting LPS-induced inflammatory response by blocking signaling STING [100]. These authors showed that S1P directly interacted with STING and reduced IFN-β generation. Thus, S1P produced by SPHK1 enhances barrier function by stimulating S1PR1 in a paracrine manner [122] while SPHK2 induce S1P appears to act in a cell-context dependent manner and modulates gene transcription, cellular proliferation and inflammatory responses independent of S1PRs receptor activation.

Several enzymes can catabolize S1P [103, 108]. S1P lyase (SPL), which is localized in the ER with the catalytic domain facing the cytosol [123], irreversibly cleaves S1P into hexadecenal and phosphoethanolamine [44, 124]. This degradation reaction represents the last step in sphingolipid catabolism. Additionally, S1P can be dephosphorylated into SPH, through two specific ER localized phosphatase namely, S1P phosphatases (SPP1 and SPP2), or through broadly-specific membrane localized lipid phosphate phosphatases (LPP1–3) [44, 124126]. SPH can either recycle back to S1P by SPHK1 or SPHK2. SPH can also be converted into ceramide by ceramide synthase.

Deficiency of SPL led to early death due to significant developmental and functional defects, such as anemia, myeloid cell hyperplasia and multiple congenital anomalies in various organs [127, 128]. SPL−/− lung alveoli showed increased number of alveolar macrophages and deposition of hyaline material [129]. These mice also showed increased egress of neutrophils and accumulation of pro-inflammatory cytokines such as TNF, INF-γ, MCP1 and IL-6 in the circulation [130]. However, Zhao et al showed that LPS enhanced SPL expression in mice [131]. Genetic or pharmacological alteration of SPL (SPL(+/-) in mice led to increased S1P levels in lung tissue and bronchoalveolar lavage fluids, and these mice showed reduced lung injury and inflammation. While the mechanisms by which SPL deficiency increased S1P levels in the lung remained enigmatic this study raise the possibility that SPPs and LPPs may compete with SPL to regulate S1P levels in EC [132]. More work is needed to clarify the divergent roles of SPL, SPPs and LPP in modulating S1P and regulation of barrier function.

S1P release from ECs is mediated by SPNS2. Spinster homolog 2 (SPNS2) is a twelve transmembrane domain protein that belongs to the SPNS/Spinster family (SPNS1–SPNS3 in vertebrates and Spinster in Drosophila). Global deletion of SPNS2 is embryonically lethal, as loss of Spns2 activity results in lethal defects in cardiovascular development [133], similar to the phenotypes of mice lacking both SPHK1 and SPHK2 or S1PR1−/− mice [20]. However, EC-specific deletion of Spns2 resulted in defects of lymphocyte egress similar to those observed in the global SPNS2-KO mice [134]. SPNS2 deletion in EC decreased circulating S1P to the same level as that of the global knockout [134, 135], and also had similar lymphopenic effects [136, 137]. SPNS2 overexpression led to increased secretion of S1P [133, 136], while down-regulation of SPNS2 reduced the release of both S1P and dihydro-S1P [136], thus proving that SPNS2 is a mammalian S1P transporter. In addition to endogenously produced dihydro-S1P and S1P, S1P analogues, such as FTY720 phosphate, can also be transported by SPNS2, but not the unphosphorylated sphingoid bases [136, 138]. Additionally, it was recently shown that a member of the same family of transporters as SPNS2, Mfsd2b, is responsible for releasing S1P from erythrocytes and platelets, and mice lacking Mfsd2b showed ~50% lower plasma levels [139]. It would be interesting to determine if loss of Mfs2b alters SPNS2, SPHK1 and SPHK2 and thereby barrier function.

Role of S1P chaperon in regulating S1PRs function

S1P binds to protein carriers such as albumin (≈35%), high-densitylipoprotein (HDL) (≈60%), and to a lesser extent to low-densityand very low-density lipoproteins [140]. HDL is known to have various atheroprotective functions, because of its anti-oxidative, anti-thrombotic, and anti-inflammatory properties [141, 142]. HDL contains various apolipoproteins including ApoA1-II, and ApoM [143]. ApoA and APoM suppress oxidative and inflammatory signaling [144, 145]. Hla’s group compared the effect of S1P bound to albumin versus ApoM in altering inflammatory signaling in endothelial cells [11, 146]. They showed that compared to albumin bound-S1P, ApoM-bound S1P had a higher half-life, served as a biased agonist for S1PR1 and inhibited ICAM1 and VCAM expression to a greater extent [11]. Mechanistically, these authors showed that ApoM-S1P promoted complex formation between S1PR1 and β-arrestin at the plasma membrane, which in turn limited endocytosis of S1PR1. In contrast, the albumin-bound S1P-S1PR1 endocytosed rapidly and induced markedly enhanced Gi-mediated signaling [147]. Consistent with these findings, loss of ApoM globally in mice resulted in ~50% lower S1P levels and induced lung vascular leak in an S1PR1-dependent manner [148, 149]. Obinata et al, using albumin-deficient mice, identified ApoA4 as another S1P binding protein, since recombinant ApoA4 activated multiple S1P receptors, and enhanced the barrier function of vascular endothelium [11]. Whether ApoM and ApoA4 have additional functions, including regulation of SPHK, sphingosine phosphate lyase and SPNS2, remain elusive.

Regulation of S1PR1

Crystal structure of S1PR1 indicates that S1PR1 shares many common features with class A GPCRs including an extracellular N-terminus, seven membrane-spanning α-helices (TM), and an intracellular C-terminus. As with other GPCRs, S1PR1 is localized on the plasma membrane of endothelial cells [47, 150, 151]. Upon ligation with S1P, S1PR1 is rapidly desensitized due to depletion of the receptor from the cell surface [47, 150, 152]. S1PR1 recycles back to the surface in the next 1–2 h following S1P binding. In contrast to S1P, the phospho-FTY720, a stable S1PR1 analog, prevents S1PR1 recycling to cell-surface by promoting S1PR1 degradation [89]. Phospho-FTY720 is generated in the cell from its prodrug FTY720 (also known as Fingolimod/Gilenya). FTY720 is phosphorylated by sphingosine kinases, predominantly SPHK2 [153]. Phospho-FTY720 is then exported possibly by Spns2 to activate S1PR1 in a paracrine manner. Thus, while FTY720 in itself is a weak agonist for S1PR1 the phosphor-FTY720 binds S1PR1 irreversibly leading to S1PR1 degradation and barrier disruption [1, 154]. Since inflammatory mediators such as LPS disrupts barrier function a possibility exist that these mediator’s function by degrading S1PR1. In this scenario, resynthesis of S1PR1 and its trafficking to the cell-surface will be required to reverse the vascular injury [155, 156]. Below we discuss S1PR1 regulation by post-translational and transcriptional mechanisms.

Overview of GPCR trafficking

GPCR signaling is regulated through agonist-induced phosphorylation of the receptor followed by receptor internalization. G-protein receptor kinases, known as GRK, phosphorylate GPCRs on their C-terminus allowing uncoupling of GPCR/G-protein signaling (desensitization) [157, 158]. This is followed by the recruitment of β-arrestin1 and β-arrestin2 proteins to phosphorylated GPCRs, thus facilitating the uncoupling of the receptor from the hetereotrimeric G-protein and endocytosis of GPCRs to endosomes to allow GPCR dephosphorylation and resensitization [159, 160]. β-arrestins bind to both the clathrin heavy chain and the β2-adaptin subunit of the heterotetrameric adaptor complex to facilitate endocytosis [161]. Clathrin, AP-2, and endocytic accessory proteins assemble at the plasma membrane to form invaginating clathrin-coated pits (CCPs). CCPs then undergo a maturation process in which the pits are pinched off to form clathrin-coated vesicles (CCVs) [162]. Clathrin-independent endocytosis, which occurs through lipid rafts and a caveolin-mediated pathway, has recently emerged as another important trafficking pathway [163]. Despite the internalization pathway, it is generally thought that internalized receptors are trafficked to an early endosome, where they are sorted for degradation, recycling, or both [159, 164]. Evidence also indicates that the interaction of β-arrestins with GPCRs determines whether GPCRs are dephosphorylated, resensitized or rapidly recycled or not recycled to the plasma membrane [161, 165].

Serine-threonine phosphorylation of S1PR1 and regulation of vascular barrier function

The C-terminal tail of GPCRs contains clusters of Ser/Thr (e.g., SSS, SXSS, SSXS) which represent a barcode for formation of stable complexes between β-arrestinand GPCR [166]. In this context, Lee et al showed that deletion of the serine rich domain within the C-terminus (SRSKSDNSS) of S1PR1 inhibited receptor internalization by S1P [5, 150], indicating that S1P induces the phosphorylation of S1PR1 at the serine rich domain. While kinases inducing S1PR1 phosphorylation at the serine rich domain were not identified, Oo et al proposed that this region is a potential phosphorylation site for casein kinase II and protein kinase C [150] (Fig. 2A).

Figure 2: S1PR1 trafficking and barrier function upon serine/tyrosine phosphorylation.

Figure 2:

S1P/phosphorylated-FTY720 (FTY720-P) activates PKC or casein kinase II (CKII) which phosphorylates S1PR1 at five-serine residues at C-terminal domain (A). Phosphorylated S1PR1 binds beta-arrestin leading to receptor internalization. In the case of S1P, serine phosphorylated receptor recycles back to cell-surface. However, FTY-720-P phosphorylated S1PR1 binds the ubiquitin ligase WWP2, which leads to degradation of receptor and loss of endothelial barrier function. B, S1P also activates pp60Src which phosphorylates S1PR1 at tyrosine 143. Y143-S1PR1 is internalized by beta-arrestin-dynamin pathway. Y143-S1PR1 recycles back to the cell-surface after transiently localizing at endoplasmic reticulum (ER) to mobilize intracellular Ca2+. However, in contrast to serine-phosphorylated S1PR1, tyrosine phosphorylated S1PR1 mutant (Y143D-S1PR1 mutant) is shunted from being degraded, persistently localizes at ER to sustain the increase in cytosolic Ca2+ in a Gi-dependent manner disrupting barrier function. Thus, finding approaches to block tyrosine phosphorylation of S1PR1 may provide novel S1PR1 barrier protective agonists.

As mentioned above, serine-phosphorylated S1PR1 was internalized primarily by the clarthrin-dependent route [167]. In Chinese hamster ovary cells coexpressing S1PR1 and β-arrestin, activation of S1PR1 led to recruitment of β-arrestin [168], which was inhibited in cells depleted of clathrin, AP-2 or β-arrestin1 and β-arrestin2 [152]. Similarly, inhibition of dynamin, a large GTPase that promotes endocytosis of clathrin-coated pits or caveolae from the plasma membrane, inhibited S1PR1 internalization [152], indicating that S1PR1 is internalized by clathrin-dependent mechanisms. However, Oo et al also showed that S1PR1 recycled back to cell-surface after 1–2 h of S1P stimulation [150]. While the phosphatase and the recycling endosomes involved in trafficking of dephosphorylated serine-phosphorylated S1PR1 remains to be determined, a report showed that internalized S1PR1 colocalizes with clathrin and an early endosome marker, Rabaptin-5, in S1P-stimulated T cells [169] (Fig. 2A).

In contrast to S1P, FTY720P was shown to induce persistent phosphorylation of the C-terminal domain of S1PR1 at S351, S353, S355, S358 and S359 [89]. These authors showed that the serine phosphorylated S1PR1 bound the E3 ubiquitin ligase WWP2. WWP2 in turn ubiquitinated serines to lysines, thus inducing S1PR1 degradation. However, related E3 ubiquitin ligases NEDD4–1, NEDD4–2, AIP4, and Cbl were not required, supporting the specificity of S1PR1 ubiquitination. S1PR1 phosphorylation induced pulmonary vascular leak, while S5A-S1PR1 mutant mice were protected (Fig. 2A). Interestingly, a phosphodefective mutant (5Ser→5Ala) of S1PR1 or an S1PR1 mutant incapable of undergoing ubiquitination at lysine residues (S1PR1-K4R mutant), bound WWP2, indicating that neither phosphorylation nor ubiquitination of the receptor were required for S1PR1 interaction with WWP2. These studies raise an important question whether WWP2 controls the cell-surface pool of S1PR1 under basal conditions.

Tyrosine phosphorylation of S1PR1 and regulation of endothelial barrier function

In addition to serine/threonine residues, S1PR1 contains 3 tyrosine residues of unknown function. We demonstrated that S1P rapidly phosphorylates S1PR1 on tyrosine residues in EC and HEK293 cells transducing S1PR1, which precedes receptor internalization. S1PR1 recycled back to the cell surface within 1 h [47]. Mutational analysis identified Y143 as the critical residue modulating S1PR1 cell surface expression. We showed that while a phospho-defective S1PR1-mutant (S1PR1-Y143F) resisted internalization, a phospho-mimicking S1PR1 mutant (S1PR1-Y143D) showed unusually high receptor internalization (Fig. 2B). Compared to WT-S1PR1-transducing endothelial cells, EC transducing the Y143F-S1PR1 mutant showed enhanced barrier function. However, barrier function was markedly decreased in EC expressing the Y143D-S1PR1 mutant, indicating that inhibition of tyrosine phosphorylation at Y143 enhanced basal endothelial barrier function. Interestingly, phosphorylation of the S1PR1 C-terminal serine residues did not alter the role of Y143 phosphorylation in signaling S1PR1 internalization.

Chavez et al, showed that S1PR1 phosphorylation at Y143 was induced by cSrc [47]. Because S1P induces the activity of Src kinases [170172], it is possible that S1PR1 itself recruits cSrc to dynamically regulate its responsiveness to S1P. Consistent with this notion, S1PR1 is shown to form a functional complex with PDGFRβ [172, 173], which by recruiting c-Src regulates S1PR1-Gi signaling.

The identity of the protein tyrosine phosphatase that dephosphorylates S1PR1 is unclear. SHP2, PTP-1B and VE-PTP all associate with adherens junctions in endothelial cells, and the proteins associated with AJ are known to be regulated by dephosphorylation [174, 175]. It is quite possible that these phosphatases induce S1PR1 dephosphorylation, thus reinstating endothelial cell-surface expression. Anwar et al showed that Y143D-S1PR1 mutant forms a complex with dynamin and inhibition of dynamin blocks the internalization of Y143D-S1PR1, leading to cell surface retention of the receptor [176]. These authors showed that internalized Y143D-S1PR1 was resident in endoplasmic reticulum (ER) and remained functional. In contrast to wild-type S1PR1, the mutated S1PR1 (Y143D-S1PR1) markedly enhanced increased intracellular Ca2+ in response to S1P in a Gi-dependent manner and disrupted EC barrier function (Fig. 2B). Thus, rapid reduction of EC surface expression of S1PR1 subsequent to serine and Y143 phosphorylation is a crucial mechanism modulating S1PR1 cell-surface expression and signaling and hence the endothelial barrier repair function of S1P. EC shows considerable heterogeneity in terms of barrier integrity in various vascular beds [3]. Whether serine versus tyrosine phosphorylation of S1PR1 occurs in a tissue specific manner to regulate their barrier property remains to parse out. Also, further studies will be required to determine why Y143D mutant localized to ER and what are its binding partners.

Transcriptional mechanisms inducing S1PR1 synthesis

Besides acute GPCR internalization-recycling signaling, evidence suggests that S1PR1 is also synthesized following EC barrier dysfunction. Simvastatin, an HMG-CoA reductase inhibitor, is a well-known anti-inflammatory and vaso-protective reagent in murine and human inflammatory diseases [177, 178]. Like S1P, simvastatin also enhanced endothelial barrier function [179]. Sun et al showed that simvastatin activates the transcriptional activity of Kruppel-like factor 2 (KLF2) [156]. Mechanism by which simvastatin induce KLF2 expression is not clear but may involve ERK activity [180, 181]. KLF2 contains a conserved myocyte enhancer factor 2c (MEF2c)-binding site [180, 181]. Evidence indicates that ERK increases the transcriptional activity of MEF2c [182]. Thus, statin by inducing ERK activity may increase KLF2 synthesis by MEF2c. KLF2 is a zinc-finger transcription factor that directly promotes expression of the genes encoding sphingosine-1 phosphate receptor 1 (S1PR1) [183185]. Consequently, EC-specific KLF2-null mice showed decreased barrier function [156, 185, 186]. We have shown that KLF2 induces S1PR1 expression downstream of focal adhesion kinase (FAK) [187, 188]. Loss of FAK in endothelium spontaneously induces vascular leak in mice, recapitulating the findings in EC-S1PR1-null mice [189]. We showed that FAK forms the endothelial barrier by maintaining S1PR1 expression. Thus, depletion of FAK significantly reduces the expression of S1PR1 both at the mRNA and protein level without changing the expression of other S1PRs [187]. Intriguingly, we found that depletion of FAK led to hypermethylation of KLF2 by DNA methylases [188].

Cai et al showed that FOXF1, the forkhead transcription factor known to regulate embryonic lung development, bound the S1PR1 promoter and thereby increased S1PR1 synthesis [155]. Thus, loss of FOXF1 in EC impairs S1PR1 synthesis, disrupting lung vascular homeostasis. Akhter et al (un-published data) also showed that S1PR1 activates STAT3, which in turn induces S1PR1 synthesis [190]. Together these studies indicate that new synthesis of S1PR1 can rescue S1PR1 cell-surface pool and thereby EC-barrier function.

Summary and Concluding Remarks

This review describes recent developments in the area of endothelial barrier function. We particularly focused on regulation of endothelial barrier function by S1PR1. As such, we describe herein the critical role of S1P generation and transport by SPHK1/SPNS2 in regulating S1PR1 activity, which in turn maintains endothelial barrier integrity. Intriguingly, data also show a key role of serine versus tyrosine phosphorylation of S1PR1 in regulating the EC-surface expression. However, several questions remain to be addressed, such as 1) how inflammatory mediators selectively alter the serine versus tyrosine phosphorylation of S1PR1 in EC?, 2) what is the pool of S1PR1 that is phosphorylated on serine versus tyrosine residues and does it occur in a vascular bed specific manner, 3) whether tyrosine-phosphorylated S1PR1 interacts differentially with barrier disrupting or barrier forming mechanisms (RhoA versus Rac1) to alter endothelial permeability?, 4) what is the fate of tyrosine phosphorylated S1PR1 in ER? and 5) would development of antagonists that prevent S1PR1 phosphorylation at Y143 be beneficial in blocking unwanted vascular leak? To address these and other questions outlined in this review it will be necessary to use a combination of physiological approaches in knockout mouse models along with cell-based studies under normal and inflammatory conditions.

Highlights.

  • Sphingosine-1-phosphate receptor-1 (S1PR1), a high affinity G-protein coupled receptor expressed at the endothelial cell (EC) surface, upon ligating its ligand S1P protects endothelial barrier by signaling adherens junction assembly.

  • Loss of S1PR1 from EC-surface disrupts endothelial barrier function leading to inflammatory conditions, such as acute lung injury and respiratory distress syndrome (ARDS).

  • Post-translational modification of S1PR1 at serine and tyrosine-143 residues play an important role in modulating functional pool of S1PR1 at the EC-surface.

  • New synthesis of S1PR1 can rescue S1PR1 EC-surface pool during injury for restoring EC-barrier function.

Acknowledgements

Funding: This work was supported by National Institutes of Health (NIH) Grants HL060678, HL084153 to D.M.

Footnotes

Conflict of Interest: None declared

Declaration of interests

The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper.

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

References

  • [1].Matloubian M, Lo CG, Cinamon G, Lesneski MJ, Xu Y, Brinkmann V, Allende ML, Proia RL, Cyster JG, Lymphocyte egress from thymus and peripheral lymphoid organs is dependent on S1P receptor 1, Nature 427(6972) (2004) 355–60. [DOI] [PubMed] [Google Scholar]
  • [2].Hla T, Physiological and pathological actions of sphingosine 1-phosphate, Semin Cell Dev Biol 15(5) (2004) 513–20. [DOI] [PubMed] [Google Scholar]
  • [3].Mehta D, Malik AB, Signaling mechanisms regulating endothelial permeability, Physiol Rev 86(1) (2006) 279–367. [DOI] [PubMed] [Google Scholar]
  • [4].Mendelson K, Zygmunt T, Torres-Vazquez J, Evans T, Hla T, Sphingosine 1-phosphate receptor signaling regulates proper embryonic vascular patterning, J Biol Chem 288(4) (2013) 2143–56. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [5].Lee MJ, Van Brocklyn JR, Thangada S, Liu CH, Hand AR, Menzeleev R, Spiegel S, Hla T, Sphingosine-1-phosphate as a ligand for the G protein-coupled receptor EDG-1, Science 279(5356) (1998) 1552–5. [DOI] [PubMed] [Google Scholar]
  • [6].Venkatakrishnan AJ, Deupi X, Lebon G, Heydenreich FM, Flock T, Miljus T, Balaji S, Bouvier M, Veprintsev DB, Tate CG, Schertler GF, Babu MM, Diverse activation pathways in class A GPCRs converge near the G-protein-coupling region, Nature 536(7617) (2016) 484–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [7].Sanchez T, Hla T, Structural and functional characteristics of S1P receptors, J Cell Biochem 92(5) (2004) 913–22. [DOI] [PubMed] [Google Scholar]
  • [8].Chun J, Goetzl EJ, Hla T, Igarashi Y, Lynch KR, Moolenaar W, Pyne S, Tigyi G, International Union of Pharmacology. XXXIV. Lysophospholipid receptor nomenclature, Pharmacol Rev 54(2) (2002) 265–9. [DOI] [PubMed] [Google Scholar]
  • [9].Morales P, Hurst DP, Reggio PH, Methods for the Development of In Silico GPCR Models, Methods Enzymol 593 (2017) 405–448. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [10].Blaho VA, Hla T, An update on the biology of sphingosine 1-phosphate receptors, J Lipid Res 55(8) (2014) 1596–608. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [11].Obinata H, Hla T, Sphingosine 1-phosphate and inflammation, International Immunology 31(9) (2019) 617–625. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [12].Baeyens A, Fang V, Chen C, Schwab SR, Exit Strategies: S1P Signaling and T Cell Migration, Trends Immunol 36(12) (2015) 778–787. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [13].Sanna MG, Liao J, Jo E, Alfonso C, Ahn MY, Peterson MS, Webb B, Lefebvre S, Chun J, Gray N, Rosen H, Sphingosine 1-phosphate (S1P) receptor subtypes S1P1 and S1P3, respectively, regulate lymphocyte recirculation and heart rate, J Biol Chem 279(14) (2004) 13839–48. [DOI] [PubMed] [Google Scholar]
  • [14].Komarova YA, Kruse K, Mehta D, Malik AB, Protein Interactions at Endothelial Junctions and Signaling Mechanisms Regulating Endothelial Permeability, Circ Res 120(1) (2017) 179–206. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [15].Predescu D, Palade GE, Plasmalemmal vesicles represent the large pore system of continuous microvascular endothelium, Am J Physiol 265(2 Pt 2) (1993) H725–33. [DOI] [PubMed] [Google Scholar]
  • [16].Minshall RD, Tiruppathi C, Vogel SM, Niles WD, Gilchrist A, Hamm HE, Malik AB, Endothelial cell-surface gp60 activates vesicle formation and trafficking via G(i)-coupled Src kinase signaling pathway, J Cell Biol 150(5) (2000) 1057–70. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [17].Garcia JG, Liu F, Verin AD, Birukova A, Dechert MA, Gerthoffer WT, Bamberg JR, English D, Sphingosine 1-phosphate promotes endothelial cell barrier integrity by Edg-dependent cytoskeletal rearrangement, J Clin Invest 108(5) (2001) 689–701. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [18].Jung B, Obinata H, Galvani S, Mendelson K, Ding BS, Skoura A, Kinzel B, Brinkmann V, Rafii S, Evans T, Hla T, Flow-regulated endothelial S1P receptor-1 signaling sustains vascular development, Dev Cell 23(3) (2012) 600–10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [19].Thennes T, Mehta D, Heterotrimeric G proteins, focal adhesion kinase, and endothelial barrier function, Microvascular research 83(1) (2012) 31–44. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [20].Liu Y, Wada R, Yamashita T, Mi Y, Deng CX, Hobson JP, Rosenfeldt HM, Nava VE, Chae SS, Lee MJ, Liu CH, Hla T, Spiegel S, Proia RL, Edg-1, the G protein-coupled receptor for sphingosine-1-phosphate, is essential for vascular maturation, J Clin Invest 106(8) (2000) 951–61. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [21].Allende ML, Yamashita T, Proia RL, G-protein-coupled receptor S1P1 acts within endothelial cells to regulate vascular maturation, Blood 102(10) (2003) 3665–7. [DOI] [PubMed] [Google Scholar]
  • [22].Gaengel K, Niaudet C, Hagikura K, Lavina B, Muhl L, Hofmann JJ, Ebarasi L, Nystrom S, Rymo S, Chen LL, Pang MF, Jin Y, Raschperger E, Roswall P, Schulte D, Benedito R, Larsson J, Hellstrom M, Fuxe J, Uhlen P, Adams R, Jakobsson L, Majumdar A, Vestweber D, Uv A, Betsholtz C, The sphingosine-1-phosphate receptor S1PR1 restricts sprouting angiogenesis by regulating the interplay between VE-cadherin and VEGFR2, Dev Cell 23(3) (2012) 587–99. [DOI] [PubMed] [Google Scholar]
  • [23].Cao J, Ehling M, Marz S, Seebach J, Tarbashevich K, Sixta T, Pitulescu ME, Werner AC, Flach B, Montanez E, Raz E, Adams RH, Schnittler H, Polarized actin and VE-cadherin dynamics regulate junctional remodelling and cell migration during sprouting angiogenesis, Nat Commun 8(1) (2017) 2210. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [24].LaMontagne K, Littlewood-Evans A, Schnell C, O'Reilly T, Wyder L, Sanchez T, Probst B, Butler J, Wood A, Liau G, Billy E, Theuer A, Hla T, Wood J, Antagonism of sphingosine-1-phosphate receptors by FTY720 inhibits angiogenesis and tumor vascularization, Cancer Res 66(1) (2006) 221–31. [DOI] [PubMed] [Google Scholar]
  • [25].Balaji Ragunathrao VA, Anwar M, Akhter MZ, Chavez A, Mao Y, Natarajan V, Lakshmikanthan S, Chrzanowska-Wodnicka M, Dudek AZ, Claesson-Welsh L, Kitajewski JK, Wary KK, Malik AB, Mehta D, Sphingosine-1-Phosphate Receptor 1 Activity Promotes Tumor Growth by Amplifying VEGF-VEGFR2 Angiogenic Signaling, Cell Rep 29(11) (2019) 3472–3487 e4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [26].Kono M, Tucker AE, Tran J, Bergner JB, Turner EM, Proia RL, Sphingosine-1-phosphate receptor 1 reporter mice reveal receptor activation sites in vivo, J Clin Invest 124(5) (2014) 2076–86. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [27].Rivera J, Proia RL, Olivera A, The alliance of sphingosine-1-phosphate and its receptors in immunity, Nat Rev Immunol 8(10) (2008) 753–63. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [28].Adada M, Canals D, Hannun YA, Obeid LM, Sphingosine-1-phosphate receptor 2, FEBS J 280(24) (2013) 6354–66. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [29].Broder S, Venter JC, Sequencing the entire genomes of free-living organisms: the foundation of pharmacology in the new millennium, Annu Rev Pharmacol Toxicol 40 (2000) 97–132. [DOI] [PubMed] [Google Scholar]
  • [30].Mandala S, Hajdu R, Bergstrom J, Quackenbush E, Xie J, Milligan J, Thornton R, Shei GJ, Card D, Keohane C, Rosenbach M, Hale J, Lynch CL, Rupprecht K, Parsons W, Rosen H, Alteration of lymphocyte trafficking by sphingosine-1-phosphate receptor agonists, Science 296(5566) (2002) 346–9. [DOI] [PubMed] [Google Scholar]
  • [31].Rosen H, Gonzalez-Cabrera PJ, Sanna MG, Brown S, Sphingosine 1-phosphate receptor signaling, Annu Rev Biochem 78 (2009) 743–68. [DOI] [PubMed] [Google Scholar]
  • [32].Allende ML, Proia RL, Sphingosine-1-phosphate receptors and the development of the vascular system, Biochim Biophys Acta 1582(1–3) (2002) 222–7. [DOI] [PubMed] [Google Scholar]
  • [33].Brinkmann V, Billich A, Baumruker T, Heining P, Schmouder R, Francis G, Aradhye S, Burtin P, Fingolimod (FTY720): discovery and development of an oral drug to treat multiple sclerosis, Nat Rev Drug Discov 9(11) (2010) 883–97. [DOI] [PubMed] [Google Scholar]
  • [34].Maceyka M, Harikumar KB, Milstien S, Spiegel S, Sphingosine-1-phosphate signaling and its role in disease, Trends Cell Biol 22(1) (2012) 50–60. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [35].Hla T, Maciag T, An abundant transcript induced in differentiating human endothelial cells encodes a polypeptide with structural similarities to G-protein-coupled receptors, J Biol Chem 265(16) (1990) 9308–13. [PubMed] [Google Scholar]
  • [36].Aarthi JJ, Darendeliler MA, Pushparaj PN, Dissecting the role of the S1P/S1PR axis in health and disease, J Dent Res 90(7) (2011) 841–54. [DOI] [PubMed] [Google Scholar]
  • [37].Spiegel S, Milstien S, The outs and the ins of sphingosine-1-phosphate in immunity, Nat Rev Immunol 11(6) (2011) 403–15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [38].Howlett AC, Reggio PH, Childers SR, Hampson RE, Ulloa NM, Deutsch DG, Endocannabinoid tone versus constitutive activity of cannabinoid receptors, Br J Pharmacol 163(7) (2011) 1329–43. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [39].Mizugishi K, Yamashita T, Olivera A, Miller GF, Spiegel S, Proia RL, Essential role for sphingosine kinases in neural and vascular development, Mol Cell Biol 25(24) (2005) 11113–21. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [40].Takabe K, Paugh SW, Milstien S, Spiegel S, "Inside-out" signaling of sphingosine-1-phosphate: therapeutic targets, Pharmacol Rev 60(2) (2008) 181–95. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [41].Rosen H, Stevens RC, Hanson M, Roberts E, Oldstone MB, Sphingosine-1-phosphate and its receptors: structure, signaling, and influence, Annu Rev Biochem 82 (2013) 637–62. [DOI] [PubMed] [Google Scholar]
  • [42].Osborne N, Brand-Arzamendi K, Ober EA, Jin SW, Verkade H, Holtzman NG, Yelon D, Stainier DY, The spinster homolog, two of hearts, is required for sphingosine 1-phosphate signaling in zebrafish, Curr Biol 18(23) (2008) 1882–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [43].Graeler M, Goetzl EJ, Activation-regulated expression and chemotactic function of sphingosine 1-phosphate receptors in mouse splenic T cells, FASEB J 16(14) (2002) 1874–8. [DOI] [PubMed] [Google Scholar]
  • [44].Fyrst H, Saba JD, An update on sphingosine-1-phosphate and other sphingolipid mediators, Nat Chem Biol 6(7) (2010) 489–97. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [45].Theilmeier G, Schmidt C, Herrmann J, Keul P, Schafers M, Herrgott I, Mersmann J, Larmann J, Hermann S, Stypmann J, Schober O, Hildebrand R, Schulz R, Heusch G, Haude M, von Wnuck Lipinski K, Herzog C, Schmitz M, Erbel R, Chun J, Levkau B, High-density lipoproteins and their constituent, sphingosine-1-phosphate, directly protect the heart against ischemia/reperfusion injury in vivo via the S1P3 lysophospholipid receptor, Circulation 114(13) (2006) 1403–9. [DOI] [PubMed] [Google Scholar]
  • [46].Walzer T, Chiossone L, Chaix J, Calver A, Carozzo C, Garrigue-Antar L, Jacques Y, Baratin M, Tomasello E, Vivier E, Natural killer cell trafficking in vivo requires a dedicated sphingosine 1-phosphate receptor, Nat Immunol 8(12) (2007) 1337–44. [DOI] [PubMed] [Google Scholar]
  • [47].Chavez A, Schmidt TT, Yazbeck P, Rajput C, Desai B, Sukriti S, Giantsos-Adams K, Knezevic N, Malik AB, Mehta D, S1PR1 Tyr143 phosphorylation downregulates endothelial cell surface S1PR1 expression and responsiveness, J Cell Sci 128(5) (2015) 878–87. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [48].McVerry BJ, Garcia JG, Endothelial cell barrier regulation by sphingosine 1-phosphate, J Cell Biochem 92(6) (2004) 1075–85. [DOI] [PubMed] [Google Scholar]
  • [49].Burg N, Swendeman S, Worgall S, Hla T, Salmon JE, Sphingosine 1-Phosphate Receptor 1 Signaling Maintains Endothelial Cell Barrier Function and Protects Against Immune Complex-Induced Vascular Injury, Arthritis Rheumatol 70(11) (2018) 1879–1889. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [50].Yamamoto R, Aoki T, Koseki H, Fukuda M, Hirose J, Tsuji K, Takizawa K, Nakamura S, Miyata H, Hamakawa N, Kasuya H, Nozaki K, Hirayama Y, Aramori I, Narumiya S, A sphingosine-1-phosphate receptor type 1 agonist, ASP4058, suppresses intracranial aneurysm through promoting endothelial integrity and blocking macrophage transmigration, Br J Pharmacol 174(13) (2017) 2085–2101. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [51].Flemming S, Burkard N, Meir M, Schick MA, Germer CT, Schlegel N, Sphingosine-1-Phosphate Receptor-1 Agonist Sew2871 Causes Severe Cardiac Side Effects and Does Not Improve Microvascular Barrier Breakdown in Sepsis, Shock 49(1) (2018) 71–81. [DOI] [PubMed] [Google Scholar]
  • [52].Li Q, Chen B, Zeng C, Fan A, Yuan Y, Guo X, Huang X, Huang Q, Differential activation of receptors and signal pathways upon stimulation by different doses of sphingosine-1-phosphate in endothelial cells, Exp Physiol 100(1) (2015) 95–107. [DOI] [PubMed] [Google Scholar]
  • [53].Mehta D, Konstantoulaki M, Ahmmed GU, Malik AB, Sphingosine 1-phosphate-induced mobilization of intracellular Ca2+ mediates rac activation and adherens junction assembly in endothelial cells, J Biol Chem 280(17) (2005) 17320–8. [DOI] [PubMed] [Google Scholar]
  • [54].Reinhard NR, Mastop M, Yin T, Wu Y, Bosma EK, Gadella TWJ Jr., Goedhart J, Hordijk PL, The balance between Galphai-Cdc42/Rac and Galpha12/13-RhoA pathways determines endothelial barrier regulation by sphingosine-1-phosphate, Mol Biol Cell 28(23) (2017) 3371–3382. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [55].Fu P, Shaaya M, Harijith A, Jacobson JR, Karginov A, Natarajan V, Sphingolipids Signaling in Lamellipodia Formation and Enhancement of Endothelial Barrier Function, Curr Top Membr 82 (2018) 1–31.. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [56].Daneshjou N, Sieracki N, van Nieuw Amerongen GP, Conway DE, Schwartz MA, Komarova YA, Malik AB, Rac1 functions as a reversible tension modulator to stabilize VE-cadherin trans-interaction, J Cell Biol 208(1) (2015) 23–32. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [57].Juettner VV, Kruse K, Dan A, Vu VH, Khan Y, Le J, Leckband D, Komarova Y, Malik AB, VE-PTP stabilizes VE-cadherin junctions and the endothelial barrier via a phosphatase-independent mechanism, J Cell Biol 218(5) (2019) 1725–1742. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [58].Siddiqui MR, Komarova YA, Vogel SM, Gao X, Bonini MG, Rajasingh J, Zhao YY, Brovkovych V, Malik AB, Caveolin-1-eNOS signaling promotes p190RhoGAP-A nitration and endothelial permeability, J Cell Biol 193(5) (2011) 841–50. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [59].Singleton PA, Chatchavalvanich S, Fu P, Xing J, Birukova AA, Fortune JA, Klibanov AM, Garcia JG, Birukov KG, Akt-mediated transactivation of the S1P1 receptor in caveolin-enriched microdomains regulates endothelial barrier enhancement by oxidized phospholipids, Circ Res 104(8) (2009) 978–86. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [60].Chen W, Pendyala S, Natarajan V, Garcia JG, Jacobson JR, Endothelial cell barrier protection by simvastatin: GTPase regulation and NADPH oxidase inhibition, Am J Physiol Lung Cell Mol Physiol 295(4) (2008) L575–83. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [61].Dudek SM, Jacobson JR, Chiang ET, Birukov KG, Wang P, Zhan X, Garcia JG, Pulmonary endothelial cell barrier enhancement by sphingosine 1-phosphate: roles for cortactin and myosin light chain kinase, J Biol Chem 279(23) (2004) 24692–700. [DOI] [PubMed] [Google Scholar]
  • [62].Inagaki HK, Ben-Tabou de-Leon S, Wong AM, Jagadish S, Ishimoto H, Barnea G, Kitamoto T, Axel R, Anderson DJ, Visualizing neuromodulation in vivo: TANGO-mapping of dopamine signaling reveals appetite control of sugar sensing, Cell 148(3) (2012) 583–95. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [63].Kono M, Conlon EG, Lux SY, Yanagida K, Hla T, Proia RL, Bioluminescence imaging of G protein-coupled receptor activation in living mice, Nat Commun 8(1) (2017) 1163. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [64].Engelbrecht E, Levesque MV, He L, Vanlandewijck M, Nitzsche A, Niazi H, Kuo A, Singh SA, Aikawa M, Holton K, Proia RL, Kono M, Pu WT, Camerer E, Betsholtz C, Hla T, Sphingosine 1-phosphate-regulated transcriptomes in heterogenous arterial and lymphatic endothelium of the aorta, Elife 9 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [65].Kihara Y, Maceyka M, Spiegel S, Chun J, Lysophospholipid receptor nomenclature review: IUPHAR Review 8, Br J Pharmacol 171(15) (2014) 3575–94. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [66].Ye D, Lin F, S1pr2/Galpha13 signaling controls myocardial migration by regulating endoderm convergence, Development 140(4) (2013) 789–99. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [67].Kim GS, Yang L, Zhang G, Zhao H, Selim M, McCullough LD, Kluk MJ, Sanchez T, Critical role of sphingosine-1-phosphate receptor-2 in the disruption of cerebrovascular integrity in experimental stroke, Nat Commun 6 (2015) 7893. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [68].Cardillo C, Kilcoyne CM, Cannon RO 3rd, Panza JA, Interactions between nitric oxide and endothelin in the regulation of vascular tone of human resistance vessels in vivo, Hypertension 35(6) (2000) 1237–41. [DOI] [PubMed] [Google Scholar]
  • [69].Lorenz JN, Arend LJ, Robitz R, Paul RJ, MacLennan AJ, Vascular dysfunction in S1P2 sphingosine 1-phosphate receptor knockout mice, Am J Physiol Regul Integr Comp Physiol 292(1) (2007) R440–6. [DOI] [PubMed] [Google Scholar]
  • [70].Santos-Cortez RL, Faridi R, Rehman AU, Lee K, Ansar M, Wang X, Morell RJ, Isaacson R, Belyantseva IA, Dai H, Acharya A, Qaiser TA, Muhammad D, Ali RA, Shams S, Hassan MJ, Shahzad S, Raza SI, Bashir ZE, Smith JD, Nickerson DA, Bamshad MJ, G. University of Washington Center for Mendelian, S. Riazuddin, W. Ahmad, T.B. Friedman, S.M. Leal, Autosomal-Recessive Hearing Impairment Due to Rare Missense Variants within S1PR2, Am J Hum Genet 98(2) (2016) 331–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [71].Cattoretti G, Mandelbaum J, Lee N, Chaves AH, Mahler AM, Chadburn A, Dalla-Favera R, Pasqualucci L, MacLennan AJ, Targeted disruption of the S1P2 sphingosine 1-phosphate receptor gene leads to diffuse large B-cell lymphoma formation, Cancer Res 69(22) (2009) 8686–92. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [72].Muppidi JR, Schmitz R, Green JA, Xiao W, Larsen AB, Braun SE, An J, Xu Y, Rosenwald A, Ott G, Gascoyne RD, Rimsza LM, Campo E, Jaffe ES, Delabie J, Smeland EB, Braziel RM, Tubbs RR, Cook JR, Weisenburger DD, Chan WC, Vaidehi N, Staudt LM, Cyster JG, Loss of signalling via Galpha13 in germinal centre B-cell-derived lymphoma, Nature 516(7530) (2014) 254–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [73].Lewis CS, Voelkel-Johnson C, Smith CD, Targeting Sphingosine Kinases for the Treatment of Cancer, Adv Cancer Res 140 (2018) 295–325. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [74].Windh RT, Lee MJ, Hla T, An S, Barr AJ, Manning DR, Differential coupling of the sphingosine 1-phosphate receptors Edg-1, Edg-3, and H218/Edg-5 to the G(i), G(q), and G(12) families of heterotrimeric G proteins, J Biol Chem 274(39) (1999) 27351–8. [DOI] [PubMed] [Google Scholar]
  • [75].Sanchez T, Skoura A, Wu MT, Casserly B, Harrington EO, Hla T, Induction of vascular permeability by the sphingosine-1-phosphate receptor-2 (S1P2R) and its downstream effectors ROCK and PTEN, Arterioscler Thromb Vasc Biol 27(6) (2007) 1312–8. [DOI] [PubMed] [Google Scholar]
  • [76].Gutbier B, Schonrock SM, Ehrler C, Haberberger R, Dietert K, Gruber AD, Kummer W, Michalick L, Kuebler WM, Hocke AC, Szymanski K, Letsiou E, Luth A, Schumacher F, Kleuser B, Mitchell TJ, Bertrams W, Schmeck B, Treue D, Klauschen F, Bauer TT, Tonnies M, Weissmann N, Hippenstiel S, Suttorp N, Witzenrath M, Group CS, Sphingosine Kinase 1 Regulates Inflammation and Contributes to Acute Lung Injury in Pneumococcal Pneumonia via the Sphingosine-1-Phosphate Receptor 2, Crit Care Med 46(3) (2018) e258–e267. [DOI] [PubMed] [Google Scholar]
  • [77].Zhang G, Yang L, Kim GS, Ryan K, Lu S, O'Donnell RK, Spokes K, Shapiro N, Aird WC, Kluk MJ, Yano K, Sanchez T, Critical role of sphingosine-1-phosphate receptor 2 (S1PR2) in acute vascular inflammation, Blood 122(3) (2013) 443–55. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [78].Ni X, Epshtein Y, Chen W, Zhou T, Xie L, Garcia JG, Jacobson JR, Interaction of integrin beta4 with S1P receptors in S1P- and HGF-induced endothelial barrier enhancement, J Cell Biochem 115(6) (2014) 1187–95. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [79].Shimizu T, De Wispelaere A, Winkler M, D'Souza T, Caylor J, Chen L, Dastvan F, Deou J, Cho A, Larena-Avellaneda A, Reidy M, Daum G, Sphingosine-1-phosphate receptor 3 promotes neointimal hyperplasia in mouse iliac-femoral arteries, Arterioscler Thromb Vasc Biol 32(4) (2012) 955–61. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [80].Keul P, Lucke S, von Wnuck Lipinski K, Bode C, Graler M, Heusch G, Levkau B, Sphingosine-1-phosphate receptor 3 promotes recruitment of monocyte/macrophages in inflammation and atherosclerosis, Circ Res 108(3) (2011) 314–23. [DOI] [PubMed] [Google Scholar]
  • [81].Sun X, Singleton PA, Letsiou E, Zhao J, Belvitch P, Sammani S, Chiang ET, Moreno-Vinasco L, Wade MS, Zhou T, Liu B, Parastatidis I, Thomson L, Ischiropoulos H, Natarajan V, Jacobson JR, Machado RF, Dudek SM, Garcia JG, Sphingosine-1-phosphate receptor-3 is a novel biomarker in acute lung injury, Am J Respir Cell Mol Biol 47(5) (2012) 628–36. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [82].Niessen F, Schaffner F, Furlan-Freguia C, Pawlinski R, Bhattacharjee G, Chun J, Derian CK, Andrade-Gordon P, Rosen H, Ruf W, Dendritic cell PAR1-S1P3 signalling couples coagulation and inflammation, Nature 452(7187) (2008) 654–8. [DOI] [PubMed] [Google Scholar]
  • [83].Contos JJ, Ye X, Sah VP, Chun J, Tandem genomic arrangement of a G protein (Gna15) and G protein-coupled receptor (s1p(4)/lp(C1)/Edg6) gene, FEBS Lett 531(1) (2002) 99–102. [DOI] [PubMed] [Google Scholar]
  • [84].Golfier S, Kondo S, Schulze T, Takeuchi T, Vassileva G, Achtman AH, Graler MH, Abbondanzo SJ, Wiekowski M, Kremmer E, Endo Y, Lira SA, Bacon KB, Lipp M, Shaping of terminal megakaryocyte differentiation and proplatelet development by sphingosine-1-phosphate receptor S1P4, FASEB J 24(12) (2010) 4701–10. [DOI] [PubMed] [Google Scholar]
  • [85].Ota H, Beutz MA, Ito M, Abe K, Oka M, McMurtry IF, S1P(4) receptor mediates S1P-induced vasoconstriction in normotensive and hypertensive rat lungs, Pulm Circ 1(3) (2011) 399–404. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [86].Olesch C, Ringel C, Brune B, Weigert A, Beyond Immune Cell Migration: The Emerging Role of the Sphingosine-1-phosphate Receptor S1PR4 as a Modulator of Innate Immune Cell Activation, Mediators Inflamm 2017 (2017) 6059203. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [87].Wang W, Graeler MH, Goetzl EJ, Type 4 sphingosine 1-phosphate G protein-coupled receptor (S1P4) transduces S1P effects on T cell proliferation and cytokine secretion without signaling migration, FASEB J 19(12) (2005) 1731–3. [DOI] [PubMed] [Google Scholar]
  • [88].Stepanovska B, Huwiler A, Targeting the S1P receptor signaling pathways as a promising approach for treatment of autoimmune and inflammatory diseases, Pharmacol Res (2019) 104170. [DOI] [PubMed] [Google Scholar]
  • [89].Oo ML, Chang SH, Thangada S, Wu MT, Rezaul K, Blaho V, Hwang SI, Han DK, Hla T, Engagement of S1P(1)-degradative mechanisms leads to vascular leak in mice, J Clin Invest 121(6) (2011) 2290–300. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [90].Novgorodov AS, El-Alwani M, Bielawski J, Obeid LM, Gudz TI, Activation of sphingosine-1-phosphate receptor S1P5 inhibits oligodendrocyte progenitor migration, FASEB J 21(7) (2007) 1503–14. [DOI] [PubMed] [Google Scholar]
  • [91].Jaillard C, Harrison S, Stankoff B, Aigrot MS, Calver AR, Duddy G, Walsh FS, Pangalos MN, Arimura N, Kaibuchi K, Zalc B, Lubetzki C, Edg8/S1P5: an oligodendroglial receptor with dual function on process retraction and cell survival, J Neurosci 25(6) (2005) 1459–69. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [92].van Doorn R, Lopes Pinheiro MA, Kooij G, Lakeman K, van het Hof B, van der Pol SM, Geerts D, van Horssen J, van der Valk P, van der Kam E, Ronken E, Reijerkerk A, de Vries HE, Sphingosine 1-phosphate receptor 5 mediates the immune quiescence of the human brain endothelial barrier, J Neuroinflammation 9 (2012) 133. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [93].Di Pardo A, Castaldo S, Amico E, Pepe G, Marracino F, Capocci L, Giovannelli A, Madonna M, van Bergeijk J, Buttari F, van der Kam E, Maglione V, Stimulation of S1PR5 with A-971432, a selective agonist, preserves blood-brain barrier integrity and exerts therapeutic effect in an animal model of Huntington's disease, Hum Mol Genet 27(14) (2018) 2490–2501. [DOI] [PubMed] [Google Scholar]
  • [94].Pyne NJ, Pyne S, Receptor tyrosine kinase-G-protein-coupled receptor signalling platforms: out of the shadow?, Trends Pharmacol Sci 32(8) (2011) 443–50. [DOI] [PubMed] [Google Scholar]
  • [95].Hait NC, Oskeritzian CA, Paugh SW, Milstien S, Spiegel S, Sphingosine kinases, sphingosine 1-phosphate, apoptosis and diseases, Biochim Biophys Acta 1758(12) (2006) 2016–26. [DOI] [PubMed] [Google Scholar]
  • [96].Gault CR, Obeid LM, Hannun YA, An overview of sphingolipid metabolism: from synthesis to breakdown, Adv Exp Med Biol 688 (2010) 1–23. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [97].Alvarez SE, Milstien S, Spiegel S, Autocrine and paracrine roles of sphingosine-1-phosphate, Trends Endocrinol Metab 18(8) (2007) 300–7. [DOI] [PubMed] [Google Scholar]
  • [98].Panneer Selvam S, De Palma RM, Oaks JJ, Oleinik N, Peterson YK, Stahelin RV, Skordalakes E, Ponnusamy S, Garrett-Mayer E, Smith CD, Ogretmen B, Binding of the sphingolipid S1P to hTERT stabilizes telomerase at the nuclear periphery by allosterically mimicking protein phosphorylation, Sci Signal 8(381) (2015) ra58. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [99].Hait NC, Allegood J, Maceyka M, Strub GM, Harikumar KB, Singh SK, Luo C, Marmorstein R, Kordula T, Milstien S, Spiegel S, Regulation of histone acetylation in the nucleus by sphingosine-1-phosphate, Science 325(5945) (2009) 1254–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [100].Joshi JC, Joshi B, Rochford I, Rayees S, Akhter MZ, Baweja S, Chava KR, Tauseef M, Abdelkarim H, Natarajan V, Gaponenko V, Mehta D, SPHK2-Generated S1P in CD11b(+) Macrophages Blocks STING to Suppress the Inflammatory Function of Alveolar Macrophages, Cell Rep 30(12) (2020) 4096–4109 e5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [101].Yatomi Y, Igarashi Y, Yang L, Hisano N, Qi R, Asazuma N, Satoh K, Ozaki Y, Kume S, Sphingosine 1-phosphate, a bioactive sphingolipid abundantly stored in platelets, is a normal constituent of human plasma and serum, J Biochem 121(5) (1997) 969–73. [DOI] [PubMed] [Google Scholar]
  • [102].Gazit SL, Mariko B, Therond P, Decouture B, Xiong Y, Couty L, Bonnin P, Baudrie V, Le Gall SM, Dizier B, Zoghdani N, Ransinan J, Hamilton JR, Gaussem P, Tharaux PL, Chun J, Coughlin SR, Bachelot-Loza C, Hla T, Ho-Tin-Noe B, Camerer E, Platelet and Erythrocyte Sources of S1P Are Redundant for Vascular Development and Homeostasis, but Both Rendered Essential After Plasma S1P Depletion in Anaphylactic Shock, Circ Res 119(8) (2016) e110–26. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [103].Maceyka M, Sankala H, Hait NC, Le Stunff H, Liu H, Toman R, Collier C, Zhang M, Satin LS, Merrill AH Jr., Milstien S, Spiegel S, SphK1 and SphK2, sphingosine kinase isoenzymes with opposing functions in sphingolipid metabolism, J Biol Chem 280(44) (2005) 37118–29. [DOI] [PubMed] [Google Scholar]
  • [104].Pitson SM, Regulation of sphingosine kinase and sphingolipid signaling, Trends Biochem Sci 36(2) (2011) 97–107. [DOI] [PubMed] [Google Scholar]
  • [105].Chan H, Pitson SM, Post-translational regulation of sphingosine kinases, Biochim Biophys Acta 1831(1) (2013) 147–56. [DOI] [PubMed] [Google Scholar]
  • [106].Liu H, Sugiura M, Nava VE, Edsall LC, Kono K, Poulton S, Milstien S, Kohama T, Spiegel S, Molecular cloning and functional characterization of a novel mammalian sphingosine kinase type 2 isoform, J Biol Chem 275(26) (2000) 19513–20. [DOI] [PubMed] [Google Scholar]
  • [107].Mendelson K, Evans T, Hla T, Sphingosine 1-phosphate signalling, Development 141(1) (2014) 5–9.. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [108].Saba JD, Hla T, Point-counterpoint of sphingosine 1-phosphate metabolism, Circ Res 94(6) (2004) 724–34. [DOI] [PubMed] [Google Scholar]
  • [109].Xiong Y, Yang P, Proia RL, Hla T, Erythrocyte-derived sphingosine 1-phosphate is essential for vascular development, J Clin Invest 124(11) (2014) 4823–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [110].Olivera A, Kohama T, Edsall L, Nava V, Cuvillier O, Poulton S, Spiegel S, Sphingosine kinase expression increases intracellular sphingosine-1-phosphate and promotes cell growth and survival, J Cell Biol 147(3) (1999) 545–58. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [111].Strub GM, Maceyka M, Hait NC, Milstien S, Spiegel S, Extracellular and intracellular actions of sphingosine-1-phosphate, Adv Exp Med Biol 688 (2010) 141–55. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [112].Venkataraman K, Thangada S, Michaud J, Oo ML, Ai Y, Lee YM, Wu M, Parikh NS, Khan F, Proia RL, Hla T, Extracellular export of sphingosine kinase-1a contributes to the vascular S1P gradient, Biochem J 397(3) (2006) 461–71. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [113].Pitson SM, Moretti PA, Zebol JR, Lynn HE, Xia P, Vadas MA, Wattenberg BW, Activation of sphingosine kinase 1 by ERK1/2-mediated phosphorylation, EMBO J 22(20) (2003) 5491–500. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [114].Pitson SM, Xia P, Leclercq TM, Moretti PA, Zebol JR, Lynn HE, Wattenberg BW, Vadas MA, Phosphorylation-dependent translocation of sphingosine kinase to the plasma membrane drives its oncogenic signalling, J Exp Med 201(1) (2005) 49–54. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [115].Shen H, Giordano F, Wu Y, Chan J, Zhu C, Milosevic I, Wu X, Yao K, Chen B, Baumgart T, Sieburth D, De Camilli P, Coupling between endocytosis and sphingosine kinase 1 recruitment, Nat Cell Biol 16(7) (2014) 652–62. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [116].Johnson KR, Becker KP, Facchinetti MM, Hannun YA, Obeid LM, PKC-dependent activation of sphingosine kinase 1 and translocation to the plasma membrane. Extracellular release of sphingosine-1-phosphate induced by phorbol 12-myristate 13-acetate (PMA), J Biol Chem 277(38) (2002) 35257–62. [DOI] [PubMed] [Google Scholar]
  • [117].Tauseef M, Kini V, Knezevic N, Brannan M, Ramchandaran R, Fyrst H, Saba J, Vogel SM, Malik AB, Mehta D, Activation of sphingosine kinase-1 reverses the increase in lung vascular permeability through sphingosine-1-phosphate receptor signaling in endothelial cells, Circ Res 103(10) (2008) 1164–72.. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [118].Kunkel GT, Maceyka M, Milstien S, Spiegel S, Targeting the sphingosine-1-phosphate axis in cancer, inflammation and beyond, Nat Rev Drug Discov 12(9) (2013) 688–702. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [119].Pyne NJ, Pyne S, Sphingosine 1-Phosphate Receptor 1 Signaling in Mammalian Cells, Molecules 22(3) (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [120].Sukocheva OA, Expansion of Sphingosine Kinase and Sphingosine-1-Phosphate Receptor Function in Normal and Cancer Cells: From Membrane Restructuring to Mediation of Estrogen Signaling and Stem Cell Programming, Int J Mol Sci 19(2) (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [121].Ebenezer DL, Berdyshev EV, Bronova IA, Liu Y, Tiruppathi C, Komarova Y, Benevolenskaya EV, Suryadevara V, Ha AW, Harijith A, Tuder RM, Natarajan V, Fu P, Pseudomonas aeruginosa stimulates nuclear sphingosine-1-phosphate generation and epigenetic regulation of lung inflammatory injury, Thorax 74(6) (2019) 579–591. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [122].Ebenezer DL, Fu P, Natarajan V, Targeting sphingosine-1-phosphate signaling in lung diseases, Pharmacol Ther 168 (2016) 143–157. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [123].Ikeda M, Kihara A, Igarashi Y, Sphingosine-1-phosphate lyase SPL is an endoplasmic reticulum-resident, integral membrane protein with the pyridoxal 5'-phosphate binding domain exposed to the cytosol, Biochem Biophys Res Commun 325(1) (2004) 338–43. [DOI] [PubMed] [Google Scholar]
  • [124].Jasinska R, Zhang QX, Pilquil C, Singh I, Xu J, Dewald J, Dillon DA, Berthiaume LG, Carman GM, Waggoner DW, Brindley DN, Lipid phosphate phosphohydrolase-1 degrades exogenous glycerolipid and sphingolipid phosphate esters, Biochem J 340 ( Pt 3) (1999) 677–86. [PMC free article] [PubMed] [Google Scholar]
  • [125].Tang X, Benesch MG, Brindley DN, Lipid phosphate phosphatases and their roles in mammalian physiology and pathology, J Lipid Res 56(11) (2015) 2048–60. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [126].Le Stunff H, Peterson C, Thornton R, Milstien S, Mandala SM, Spiegel S, Characterization of murine sphingosine-1-phosphate phosphohydrolase, J Biol Chem 277(11) (2002) 8920–7. [DOI] [PubMed] [Google Scholar]
  • [127].Hagen N, Van Veldhoven PP, Proia RL, Park H, Merrill AH Jr., van Echten-Deckert G, Subcellular origin of sphingosine 1-phosphate is essential for its toxic effect in lyase-deficient neurons, J Biol Chem 284(17) (2009) 11346–53. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [128].Bektas M, Allende ML, Lee BG, Chen W, Amar MJ, Remaley AT, Saba JD, Proia RL, Sphingosine 1-phosphate lyase deficiency disrupts lipid homeostasis in liver, J Biol Chem 285(14) (2010) 10880–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [129].Vogel P, Donoviel MS, Read R, Hansen GM, Hazlewood J, Anderson SJ, Sun W, Swaffield J, Oravecz T, Incomplete inhibition of sphingosine 1-phosphate lyase modulates immune system function yet prevents early lethality and non-lymphoid lesions, PLoS One 4(1) (2009) e4112. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [130].Allende ML, Bektas M, Lee BG, Bonifacino E, Kang J, Tuymetova G, Chen W, Saba JD, Proia RL, Sphingosine-1-phosphate lyase deficiency produces a pro-inflammatory response while impairing neutrophil trafficking, J Biol Chem 286(9) (2011) 7348–58. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [131].Zhao Y, Gorshkova IA, Berdyshev E, He D, Fu P, Ma W, Su Y, Usatyuk PV, Pendyala S, Oskouian B, Saba JD, Garcia JG, Natarajan V, Protection of LPS-induced murine acute lung injury by sphingosine-1-phosphate lyase suppression, Am J Respir Cell Mol Biol 45(2) (2011) 426–35. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [132].Siow DL, Anderson CD, Berdyshev EV, Skobeleva A, Pitson SM, Wattenberg BW, Intracellular localization of sphingosine kinase 1 alters access to substrate pools but does not affect the degradative fate of sphingosine-1-phosphate, J Lipid Res 51(9) (2010) 2546–59. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [133].Kawahara A, Nishi T, Hisano Y, Fukui H, Yamaguchi A, Mochizuki N, The sphingolipid transporter spns2 functions in migration of zebrafish myocardial precursors, Science 323(5913) (2009) 524–7. [DOI] [PubMed] [Google Scholar]
  • [134].Fukuhara S, Simmons S, Kawamura S, Inoue A, Orba Y, Tokudome T, Sunden Y, Arai Y, Moriwaki K, Ishida J, Uemura A, Kiyonari H, Abe T, Fukamizu A, Hirashima M, Sawa H, Aoki J, Ishii M, Mochizuki N, The sphingosine-1-phosphate transporter Spns2 expressed on endothelial cells regulates lymphocyte trafficking in mice, J Clin Invest 122(4) (2012) 1416–26. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [135].Hisano Y, Kobayashi N, Yamaguchi A, Nishi T, Mouse SPNS2 functions as a sphingosine-1-phosphate transporter in vascular endothelial cells, PLoS One 7(6) (2012) e38941. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [136].Nagahashi M, Kim EY, Yamada A, Ramachandran S, Allegood JC, Hait NC, Maceyka M, Milstien S, Takabe K, Spiegel S, Spns2, a transporter of phosphorylated sphingoid bases, regulates their blood and lymph levels, and the lymphatic network, FASEB J 27(3) (2013) 1001–11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [137].Donoviel MS, Hait NC, Ramachandran S, Maceyka M, Takabe K, Milstien S, Oravecz T, Spiegel S, Spinster 2, a sphingosine-1-phosphate transporter, plays a critical role in inflammatory and autoimmune diseases, FASEB J 29(12) (2015) 5018–28. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [138].Hisano Y, Kobayashi N, Kawahara A, Yamaguchi A, Nishi T, The sphingosine 1-phosphate transporter, SPNS2, functions as a transporter of the phosphorylated form of the immunomodulating agent FTY720, J Biol Chem 286(3) (2011) 1758–66. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [139].Vu TM, Ishizu AN, Foo JC, Toh XR, Zhang F, Whee DM, Torta F, Cazenave-Gassiot A, Matsumura T, Kim S, Toh SES, Suda T, Silver DL, Wenk MR, Nguyen LN, Mfsd2b is essential for the sphingosine-1-phosphate export in erythrocytes and platelets, Nature 550(7677) (2017) 524–528. [DOI] [PubMed] [Google Scholar]
  • [140].Murata N, Sato K, Kon J, Tomura H, Yanagita M, Kuwabara A, Ui M, Okajima F, Interaction of sphingosine 1-phosphate with plasma components, including lipoproteins, regulates the lipid receptor-mediated actions, Biochem J 352 Pt 3 (2000) 809–15. [PMC free article] [PubMed] [Google Scholar]
  • [141].Van Lenten BJ, Hama SY, de Beer FC, Stafforini DM, McIntyre TM, Prescott SM, La Du BN, Fogelman AM, Navab M, Anti-inflammatory HDL becomes pro-inflammatory during the acute phase response. Loss of protective effect of HDL against LDL oxidation in aortic wall cell cocultures, J Clin Invest 96(6) (1995) 2758–67. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [142].Estrada-Luna D, Ortiz-Rodriguez MA, Medina-Briseno L, Carreon-Torres E, Izquierdo-Vega JA, Sharma A, Cancino-Diaz JC, Perez-Mendez O, Belefant-Miller H, Betanzos-Cabrera G, Current Therapies Focused on High-Density Lipoproteins Associated with Cardiovascular Disease, Molecules 23(11) (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [143].He L, Wu P, Tan L, Le B, Du W, Shen T, Wu J, Xiang Z, Hu M, Characteristics of lipid metabolism including serum apolipoprotein M levels in patients with primary nephrotic syndrome, Lipids Health Dis 16(1) (2017) 167. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [144].Wang M, Luo GH, Liu H, Zhang YP, Wang B, Di DM, Zhan XH, Yu Y, Yao S, Zhang XY, Xu N, Apolipoprotein M induces inhibition of inflammatory responses via the S1PR1 and DHCR24 pathways, Mol Med Rep 19(2) (2019) 1272–1283. [DOI] [PubMed] [Google Scholar]
  • [145].Zhu X, Parks JS, New roles of HDL in inflammation and hematopoiesis, Annu Rev Nutr 32 (2012) 161–82. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [146].Swendeman SL, Xiong Y, Cantalupo A, Yuan H, Burg N, Hisano Y, Cartier A, Liu CH, Engelbrecht E, Blaho V, Zhang Y, Yanagida K, Galvani S, Obinata H, Salmon JE, Sanchez T, Di Lorenzo A, Hla T, An engineered S1P chaperone attenuates hypertension and ischemic injury, Sci Signal 10(492) (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [147].Galvani S, Sanson M, Blaho VA, Swendeman SL, Obinata H, Conger H, Dahlback B, Kono M, Proia RL, Smith JD, Hla T, HDL-bound sphingosine 1-phosphate acts as a biased agonist for the endothelial cell receptor S1P1 to limit vascular inflammation, Sci Signal 8(389) (2015) ra79. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [148].Christoffersen C, Obinata H, Kumaraswamy SB, Galvani S, Ahnstrom J, Sevvana M, Egerer-Sieber C, Muller YA, Hla T, Nielsen LB, Dahlback B, Endothelium-protective sphingosine-1-phosphate provided by HDL-associated apolipoprotein M, Proc Natl Acad Sci U S A 108(23) (2011) 9613–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [149].Christensen PM, Liu CH, Swendeman SL, Obinata H, Qvortrup K, Nielsen LB, Hla T, Di Lorenzo A, Christoffersen C, Impaired endothelial barrier function in apolipoprotein M-deficient mice is dependent on sphingosine-1-phosphate receptor 1, FASEB J 30(6) (2016) 2351–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [150].Oo ML, Thangada S, Wu MT, Liu CH, Macdonald TL, Lynch KR, Lin CY, Hla T, Immunosuppressive and anti-angiogenic sphingosine 1-phosphate receptor-1 agonists induce ubiquitinylation and proteasomal degradation of the receptor, J Biol Chem 282(12) (2007) 9082–9. [DOI] [PubMed] [Google Scholar]
  • [151].Rosenbaum DM, Rasmussen SG, Kobilka BK, The structure and function of G-protein-coupled receptors, Nature 459(7245) (2009) 356–63. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [152].Reeves PM, Kang YL, Kirchhausen T, Endocytosis of Ligand-Activated Sphingosine 1-Phosphate Receptor 1 Mediated by the Clathrin-Pathway, Traffic 17(1) (2016) 40–52. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [153].Paugh SW, Payne SG, Barbour SE, Milstien S, Spiegel S, The immunosuppressant FTY720 is phosphorylated by sphingosine kinase type 2, FEBS Lett 554(1–2) (2003) 189–93. [DOI] [PubMed] [Google Scholar]
  • [154].Natarajan V, Dudek SM, Jacobson JR, Moreno-Vinasco L, Huang LS, Abassi T, Mathew B, Zhao Y, Wang L, Bittman R, Weichselbaum R, Berdyshev E, Garcia JG, Sphingosine-1-phosphate, FTY720, and sphingosine-1-phosphate receptors in the pathobiology of acute lung injury, Am J Respir Cell Mol Biol 49(1) (2013) 6–17. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [155].Cai Y, Bolte C, Le T, Goda C, Xu Y, Kalin TV, Kalinichenko VV, FOXF1 maintains endothelial barrier function and prevents edema after lung injury, Sci Signal 9(424) (2016) ra40. [DOI] [PubMed] [Google Scholar]
  • [156].Sun X, Mathew B, Sammani S, Jacobson JR, Garcia JGN, Simvastatin-induced sphingosine 1-phosphate receptor 1 expression is KLF2-dependent in human lung endothelial cells, Pulm Circ 7(1) (2017) 117–125. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [157].Tobin AB, G-protein-coupled receptor phosphorylation: where, when and by whom, Br J Pharmacol 153 Suppl 1 (2008) S167–76. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [158].Gurevich VV, Gurevich EV, GPCR Signaling Regulation: The Role of GRKs and Arrestins, Front Pharmacol 10 (2019) 125. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [159].Oakley RH, Laporte SA, Holt JA, Barak LS, Caron MG, Association of beta-arrestin with G protein-coupled receptors during clathrin-mediated endocytosis dictates the profile of receptor resensitization, J Biol Chem 274(45) (1999) 32248–57. [DOI] [PubMed] [Google Scholar]
  • [160].Beautrait A, Paradis JS, Zimmerman B, Giubilaro J, Nikolajev L, Armando S, Kobayashi H, Yamani L, Namkung Y, Heydenreich FM, Khoury E, Audet M, Roux PP, Veprintsev DB, Laporte SA, Bouvier M, A new inhibitor of the beta-arrestin/AP2 endocytic complex reveals interplay between GPCR internalization and signalling, Nat Commun 8 (2017) 15054. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [161].Tian X, Kang DS, Benovic JL, beta-arrestins and G protein-coupled receptor trafficking, Handb Exp Pharmacol 219 (2014) 173–86. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [162].McMahon HT, Boucrot E, Molecular mechanism and physiological functions of clathrin-mediated endocytosis, Nat Rev Mol Cell Biol 12(8) (2011) 517–33. [DOI] [PubMed] [Google Scholar]
  • [163].Hansen CG, Nichols BJ, Molecular mechanisms of clathrin-independent endocytosis, J Cell Sci 122(Pt 11) (2009) 1713–21. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [164].Oakley RH, Laporte SA, Holt JA, Caron MG, Barak LS, Differential affinities of visual arrestin, beta arrestin1, and beta arrestin2 for G protein-coupled receptors delineate two major classes of receptors, J Biol Chem 275(22) (2000) 17201–10. [DOI] [PubMed] [Google Scholar]
  • [165].Peterson YK, Luttrell LM, The Diverse Roles of Arrestin Scaffolds in G Protein-Coupled Receptor Signaling, Pharmacol Rev 69(3) (2017) 256–297. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [166].Oakley RH, Laporte SA, Holt JA, Barak LS, Caron MG, Molecular determinants underlying the formation of stable intracellular G protein-coupled receptor-beta-arrestin complexes after receptor endocytosis*, J Biol Chem 276(22) (2001) 19452–60. [DOI] [PubMed] [Google Scholar]
  • [167].Scarselli M, Donaldson JG, Constitutive internalization of G protein-coupled receptors and G proteins via clathrin-independent endocytosis, J Biol Chem 284(6) (2009) 3577–85. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [168].Birker-Robaczewska M, Bolli M, Rey M, de Kanter R, Kohl C, Lescop C, Boucher M, Poirey S, Steiner B, Nayler O, S1P1 Modulator-Induced G alphai Signaling and beta-Arrestin Recruitment Are Both Necessary to Induce Rapid and Efficient Reduction of Blood Lymphocyte Count In Vivo, Mol Pharmacol 93(2) (2018) 109–118. [DOI] [PubMed] [Google Scholar]
  • [169].Nomachi A, Yoshinaga M, Liu J, Kanchanawong P, Tohyama K, Thumkeo D, Watanabe T, Narumiya S, Hirata T, Moesin controls clathrin-mediated S1PR1 internalization in T cells, PLoS One 8(12) (2013) e82590. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [170].Belvitch P, Dudek SM, Role of FAK in S1P-regulated endothelial permeability, Microvasc Res 83(1) (2012) 22–30. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [171].Bergelin N, Lof C, Balthasar S, Kalhori V, Tornquist K, S1P1 and VEGFR-2 form a signaling complex with extracellularly regulated kinase 1/2 and protein kinase C-alpha regulating ML-1 thyroid carcinoma cell migration, Endocrinology 151(7) (2010) 2994–3005. [DOI] [PubMed] [Google Scholar]
  • [172].Waters CM, Connell MC, Pyne S, Pyne NJ, c-Src is involved in regulating signal transmission from PDGFbeta receptor-GPCR(s) complexes in mammalian cells, Cell Signal 17(2) (2005) 263–77. [DOI] [PubMed] [Google Scholar]
  • [173].Long JS, Natarajan V, Tigyi G, Pyne S, Pyne NJ, The functional PDGFbeta receptor-S1P1 receptor signaling complex is involved in regulating migration of mouse embryonic fibroblasts in response to platelet derived growth factor, Prostaglandins Other Lipid Mediat 80(1–2) (2006) 74–80. [DOI] [PubMed] [Google Scholar]
  • [174].Nottebaum AF, Cagna G, Winderlich M, Gamp AC, Linnepe R, Polaschegg C, Filippova K, Lyck R, Engelhardt B, Kamenyeva O, Bixel MG, Butz S, Vestweber D, VE-PTP maintains the endothelial barrier via plakoglobin and becomes dissociated from VE-cadherin by leukocytes and by VEGF, J Exp Med 205(12) (2008) 2929–45. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [175].Timmerman I, Hoogenboezem M, Bennett AM, Geerts D, Hordijk PL, van Buul JD, The tyrosine phosphatase SHP2 regulates recovery of endothelial adherens junctions through control of beta-catenin phosphorylation, Mol Biol Cell 23(21) (2012) 4212–25. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [176].Anwar M, Amin MR, Mehta D, Dynamin internalizes tyrosine phosphorylated sphingosine 1 phosphate receptor 1 and impair downstream signaling, The FASEB Journal 32(1_supplement) (2018) 557.13–557.13. [Google Scholar]
  • [177].Sparrow CP, Burton CA, Hernandez M, Mundt S, Hassing H, Patel S, Rosa R, Hermanowski-Vosatka A, Wang PR, Zhang D, Peterson L, Detmers PA, Chao YS, Wright SD, Simvastatin has anti-inflammatory and antiatherosclerotic activities independent of plasma cholesterol lowering, Arterioscler Thromb Vasc Biol 21(1) (2001) 115–21. [DOI] [PubMed] [Google Scholar]
  • [178].Mihos CG, Pineda AM, Santana O, Cardiovascular effects of statins, beyond lipid-lowering properties, Pharmacol Res 88 (2014) 12–9. [DOI] [PubMed] [Google Scholar]
  • [179].van Nieuw Amerongen GP, Beckers CM, Achekar ID, Zeeman S, Musters RJ, van Hinsbergh VW, Involvement of Rho kinase in endothelial barrier maintenance, Arterioscler Thromb Vasc Biol 27(11) (2007) 2332–9. [DOI] [PubMed] [Google Scholar]
  • [180].Sen-Banerjee S, Mir S, Lin Z, Hamik A, Atkins GB, Das H, Banerjee P, Kumar A, Jain MK, Kruppel-like factor 2 as a novel mediator of statin effects in endothelial cells, Circulation 112(5) (2005) 720–6. [DOI] [PubMed] [Google Scholar]
  • [181].Sweet DR, Fan L, Hsieh PN, Jain MK, Kruppel-Like Factors in Vascular Inflammation: Mechanistic Insights and Therapeutic Potential, Front Cardiovasc Med 5 (2018) 6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [182].Parmar KM, Larman HB, Dai G, Zhang Y, Wang ET, Moorthy SN, Kratz JR, Lin Z, Jain MK, Gimbrone MA Jr., Garcia-Cardena G, Integration of flow-dependent endothelial phenotypes by Kruppel-like factor 2, J Clin Invest 116(1) (2006) 49–58. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [183].Carlson CM, Endrizzi BT, Wu J, Ding X, Weinreich MA, Walsh ER, Wani MA, Lingrel JB, Hogquist KA, Jameson SC, Kruppel-like factor 2 regulates thymocyte and T-cell migration, Nature 442(7100) (2006) 299–302. [DOI] [PubMed] [Google Scholar]
  • [184].Skon CN, Lee JY, Anderson KG, Masopust D, Hogquist KA, Jameson SC, Transcriptional downregulation of S1pr1 is required for the establishment of resident memory CD8+ T cells, Nat Immunol 14(12) (2013) 1285–93. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [185].Sangwung P, Zhou G, Nayak L, Chan ER, Kumar S, Kang DW, Zhang R, Liao X, Lu Y, Sugi K, Fujioka H, Shi H, Lapping SD, Ghosh CC, Higgins SJ, Parikh SM, Jo H, Jain MK, KLF2 and KLF4 control endothelial identity and vascular integrity, JCI Insight 2(4) (2017) e91700. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [186].Cowan CE, Kohler EE, Dugan TA, Mirza MK, Malik AB, Wary KK, Kruppel-like factor-4 transcriptionally regulates VE-cadherin expression and endothelial barrier function, Circ Res 107(8) (2010) 959–66. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [187].Yazbeck P, Mehta D, Endothelial Focal Adhesion Kinase Transcriptionally Regulates S1P1 to Maintain Lung Vascular Barrier Function, The FASEB Journal 29(1_supplement) (2015) 625.4. [Google Scholar]
  • [188].Akhter MZ, Yazbeck P, Srivastava NN, Tauseef M, Tennes-Schmidt T, Lenzini SB, Shin J-W, Mehta D, FAK maintenance of endothelial mechanotransduction controls epigenetic repression of KLF2 and S1PR1 transcription, The FASEB Journal 32(1_supplement) (2018) 837.7–837.7. [Google Scholar]
  • [189].Schmidt TT, Tauseef M, Yue L, Bonini MG, Gothert J, Shen TL, Guan JL, Predescu S, Sadikot R, Mehta D, Conditional deletion of FAK in mice endothelium disrupts lung vascular barrier function due to destabilization of RhoA and Rac1 activities, Am J Physiol Lung Cell Mol Physiol 305(4) (2013) L291–300. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [190].Lee H, Deng J, Kujawski M, Yang C, Liu Y, Herrmann A, Kortylewski M, Horne D, Somlo G, Forman S, Jove R, Yu H, STAT3-induced S1PR1 expression is crucial for persistent STAT3 activation in tumors, Nat Med 16(12) (2010) 1421–8. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES