1. Introduction
The moniker lysophospholipid incorporates two broad families of membrane-derived lipids: the glycerophospholipids and the sphingolipids1. Primary representatives of these two arms are lysophosphatidic acid (LPA) and sphingosine 1-phosphate (S1P), the majority of whose activities are mediated via multiple G-protein-coupled receptors with a high degree of specificity for either LPA or S1P 2,3. Unlike S1P, which is a single molecular species (2S-amino-1-(dihydrogen phosphate)-4E-octadecene-1,3R-diol), LPA (1-O-acyl-2-hydroxy-sn-glycero-3-phosphate) is actually a diverse group of molecules consisting of either a saturated (e.g. 16:0; 18:0) or unsaturated (e.g. 16:1; 18:1; 18:2; 20:4) fatty acid chain esterified at the sn-1 or sn-2 position of a glycerol backbone 4,5. This can also be modified to alkyl or alkenyl at the sn-1 position 6. Other bioactive members of the LP family include the LPA analogues cyclic phosphatidic acid (cPA), sphigosylphosphotidylcholine (SPC), lysophosphatidylcholine (LPC), lysophosphatidylserine (LPS), and lysophosphatidylinositol (LPI) 1,5,7. Numerous emerging reports indicate roles in mammalian biology for other, less well-characterized LP members, as well 8. In the past decade, our understanding of LP biology has expanded exponentially, fueled by the identification of LP receptors, generation and analysis of LPA and S1P-receptor knockout mice, and small molecule compounds functioning as receptor-specific agonists or antagonists 3,9,10.
Specific receptors for LPA and S1P were cloned and characterized beginning only about 15 years ago. The description of the first LPAR was reported in 1996, and the orphan receptor EDG-1, which was cloned as an immediate-early gene from endothelial cells, was identified as the first S1P receptor in 1998 11-13. Since then, four more S1P receptors and at least five more LPA receptors have been identified 3. The two families have been best characterized in specific organ systems; namely, LPARs in the nervous system and cancer, and S1PR in vascular biology and immunology. That is not to say that their contributions in other fields are minor, only that they are incompletely understood.
2. Synthesis and degradation of LPs
The synthesis of LPA and S1P occurs both intracellularly and extracellularly 14-16,17. S1P and LPA can be found in high nanomolar to low micromolar concentrations in the blood and lymph and low concentrations in normal tissue 18-20. This LP gradient is utilized by some cell types, especially hematopoietic/immune cells, as a migratory cue and will described later.
A significant fraction of the plasma and lymph concentrations of both S1P and LPA have been attributed to production by erythrocytes and platelets, respectively 21-25. Vascular and lymphatic endothelial cells also contribute to the production of plasma and lymph S1P, respectively, and neural cells and adipocytes significant quantities of LPA 26-29. Both LPA and S1P can also be found in ovarian cancer ascites at 5-to 10-fold the typical plasma concentration, although the specific cell types responsible for this production are unknown 30,31. Synthesis of LPs by neutrophils, mast cells, mononuclear cells, intestinal, lung, and uterine epithelium, testis, ovaries, and kidneys has also been reported 32-39. It is likely the production by these, and other as yet unidentified cell types, is responsible for managing the recruitment of specific cells to sites in the microenvironment within certain tissue structures, or to stimulate growth in isolated areas, such as the LPA-mediated neurogenesis of post-mitotic cortical neurons 40. Tissue-specific knockouts of the synthetic enzymes and gene silencing studies should help to expand this list or to confirm its exclusivity.
2.1 Synthetic enzymes- autotaxin, and sphingosine kinases
Lysophopholipase D, also referred to as autotaxin (ATX), is a secreted enzyme Phospholipase A1 and A2 are responsible for the de novo synthesis of the precursor LPC 41. Deletion of the ATX gene is embryonic lethal in the mouse at e 8.5-9.5 18,42. The production of S1P from sphingosine is mediated by two kinases, Sphk1 and 2 (Figure 2) 43,44. Mice carrying deletions of only Sphk1 or 2 do not have gross abnormalities, but global deletion of both isoforms results in embryonic lethality at e10.5, similar to ATX knockouts, implying that in single knockouts, compensatory mechanisms exist 45. Additionally, no S1P was detected in Sphk1/Sphk2 double-knockout embryos, suggesting that there are no alternative pathways for S1P synthesis, at least at the embryonic development stage. Studies have utilized “triple allele” knockouts of Sphk1 and 2, where both copies of one kinase and one copy of the other kinase have been deleted, allowing for some analyses to be performed in a partial loss-of-function scenario, as well as studies using knockouts of either isoform 46-50.
Figure 2. Metabolism of S1P, S1PR G-protein coupling, and down-stream signaling pathways.

SMase, sphingomyelinase; SMS, sphingomyelin synthase; CDase, ceramidase; CerS, ceramide synthase; Sphk1/2, sphingosine kinase 1 and 2; SPP, sphingosine 1-phosphate phosphatase; LPP, lipid phosphate phosphatase; S1P, sphingosine 1-phosphate; Spns2, spinster 2 protein; ApoM, apolipoprotein M; ROCK, Rho-associated kinase; PTEN, phosphatase and tensin homolog; PLC, phospholipase C; PKC, protein kinase C; PI3K, phosphoinositide 3-kinase; MAPK, mitogen-activated protein kinase; eNOS, endothelial nitric oxide synthase.
2.2 Degradative mechanisms
Degradation of either LPA or S1P can occur in either a reversible or irreversible fashion. Dephosphorylation is mediated by a number of lipid phosphate phosphohydrolases (LPPs), all of which are integral membrane proteins, but one of which, LPP1, localizes with its active site facing the extracellular space 51,52. This indicates the possibility that it functions to dephosphorylate extracellular LPA or S1P close to membrane receptors, and thereby regulate signaling 52. For S1P, this results in conversion to sphingosine, which can then be converted back to S1P. LPA, however, is irreversibly dephosphorylated to monoacylglycerol. What could be termed “reversible degradation” of LPA occurs via lysophosphatidic acid acyltransferase beta (AGPAT2), which converts LPA to PA, which can then reenter the lipid cycle to be converted to the LPA precursor LPC53. Irreversible degradation of S1P can occur intracellularly by the lysophospholipase S1P lyase (SPL), an enzyme found in every mammalian tissue examined thus far, with the exception of platelets and erythrocytes 54-56. In many immune cells, stimulation with endotoxin can modulate the expression of multiple lysophospholipid metabolizing enzymes, including SPP, Sphk1, Sphk2, SPL 57.
3. Receptor-ligand affinities and extracellular transport
Although collectively known as lysophospholipids, the S1PR and LPAR ligand recognition is highly specific, with dissociation constants (Kd) in the nanomolar range 58,59. Activation can occur across family lines; for example, LPA can activate S1P1, but with a Kd one to two logs greater than that of S1P and thus unlikely to be physiologically relevant60,61. This low level promiscuity likely reflects the shared ancestry of these divergent receptors 62-64.
If synthesized intracellularly, how is S1P transported to the extracellular environment to exert autocrine and paracrine effects? In vitro studies using rat and human mast cell lines postulated that the ATP-binding cassette transporter protein ABCC1 was responsible for the transport of S1P from the site of synthesis in the cytoplasm or endoplasmic reticulum, through the membrane, and into the extracellular space where it could exert its influence via the GPCRs 65. Further in vitro studies also pointed to ABCA7 as a transporter in platelets and erythrocytes 66,67. However, although studies using pharmacological antagonists of either ABCA1 (glyburide) or ABCC1 (MK571) demonstrated significant suppression of endogenous S1P release, in vivo studies utilizing ABCA1-, ABCA7-, or ABCC1-null mice failed to demonstrate decreases in either plasma or lymph S1P concentrations 68.
It is important to note that such conclusions are made largely from the use of chemical inhibitors which are not specific at the doses employed 69,70. Importantly, the Spinster 2 (Spns2) protein was identified in zebrafish as an S1P transporter necessary for S1P-mediated migration of myocardial precursors 71. Recently, it was shown that human Spns2 transfected into CHO cells could transport S1P and other phosphosphingolipids (e.g. DH-S1P, phyto-S1P), as well as FTY720-P, making it the first transporter shown to have such activity 72. In the same study, Hisano, et al. demonstrated the inability of ABCA1, ABCB1, ABCC1, or ABCG2 to effectively transport S1P or FTY-P from cells over-expressing each transporter. Several possibilities exist for these conflicting data. In vivo, compensatory mechanisms, by these or other, as of yet unknown, transporters might exist that are not otherwise evident in an in vitro, single-cell system. Similarly, results for different transporters could be cell-type specific 73,74. Additionally, data obtained using MK-571 as an inhibitor of ABCC1 are unreliable, as its ability to antagonize cysteinyl leukotriene receptors and the downstream signaling pathways (e.g., calcium flux, cellular proliferation) is evident at concentrations one to two logs below the typical concentration used to block ABCC1 transport activity, a fact particularly relevant in mast cell studies 75,76. Thus far, Spns2 is the only bona fide S1P transporter, as demonstrated by both in vitro and in vivo studies 77.
4. Chaperones and carriers
Much effort has focused on determining how these lipids can arrive at the appropriate cell at the appropriate time to initiate specific signaling sequences. Lipoproteins such as high-density lipoprotein (HDL) or low-density lipoprotein (LDL) are known to be critically important for modulating vascular homeostasis 78. Whereas oxidized LDL stimulates platelet and endothelium activation and can have cytotoxic effects, HDL exerts cytoprotective and anti-atherogenic effects 78-80. Several studies using aortae from pigs, rabbits, or endothelial cell culture demonstrated that the critical component mediating this cytotoxic effect was LPC in oxLDL (the precursor of LPA) activating a GPCR81-83. Conversely, the vasoprotective qualities of HDL were not due solely to it's ability to induce cholesterol efflux from cells, but also the presence of high concentrations of S1P found bound in HDL 84-86. In fact, HDL-bound S1P accounts for approximately 65% of plasma S1P, but it was unknown how S1P was bound to this carrier, since HDL is composed of several apolipoproteins, as well as phospho- and other lipids 87.
4.1 S1P and ApoM
A recent study has shown that the apolipoprotein (apo) M is the HDL constituent responsible for the transport of S1P 88. HDL from mice lacking apoM contained no S1P, whereas over-expressing apoM resulted in higher concentrations of bound S1P that was not a result of increased HDL-cholesterol or other HDL components, as these were not significantly affected. In vitro internalization studies using cells transfected with GFP-tagged S1P1 illustrated the ability of apoM-bound S1P to activate downstream signaling cascades, whereas functional assays of endothelial cell migration and adherens junction formation determined that S1P in complex with either apoM alone or HDL was necessary for full cellular responses. Finally, an in vivo assay of vascular barrier function confirmed this, finding that mice lacking apoM demonstrated increased vascular leakage as compared to wild-type controls. This result mirrors those in plasma S1P-deficient mice and supports the idea that apoM-, not albumin-bound S1P is the necessary functional pairing for vascular barrier maintenance in vivo 21. As to how apoM mediates delivery of S1P to S1P1, and possibly to other receptors is not known, the most likely scenario involves membrane docking of the HDL particle, either directly or via binding of scavenger receptor class B type 1 (SR-B1)89. Additionally, how and where S1P is loaded onto apoM is unknown, since many cells posses the capacity for S1P synthesis. Future studies are needed to explore the transfer mechanism and it's specificity with regard to different S1P receptors.
4.2 LPA and autotaxin
Autotaxin's property as a transporter is ancillary to its enzymatic activity in the biosynthesis of LPA. In vitro binding assays showed ATX binding to lymphocytes or activated platelets in an integrin-dependent fashion, and in both instances, ATX was found in association with these cells in vivo, in high endothelial venules or thrombi, respectively 14,90. Both of these cell types respond to LPA and express multiple LPAR, making it likely that ATX is able to bind numerous cell types via integrin associations 91. Nishimasu, et al. state that the crystal structure of ATX suggests the intimate association of the ATX hydrophobic channel with the plasma membrane, subsequently allowing for the directed capture of LPA by it's receptors on the cell surface 92. It is not known if the ATX-LPA-LPA receptor association allows for cell- or receptor subtype-specific delivery of agonist, and if so, what its involvement in homeostasis or disease pathogenesis may be, and how might it be manipulated.
5. Lymphatic and cardiovascular systems
Effects on the vasculature often “read-out” as effects in other systems. Changes in vessels, large and small, can have far-reaching effects throughout an organism. Thus, to a great extent, the S1PR and LPAR can collectively modulate a multitude of responses just by altering vasculature. In these systems, the most characterized is the role of S1P1 in barrier function and vessel maturation.
5.1 S1P receptors
S1P1, originally named Edg1, was cloned from phorbol 12-myristate 12-acetate (PMA)-stimulated HUVEC in the search for immediate early genes regulating endothelial cell (EC) differentiation, thus the name “endothelial differentiation gene” 12. The importance of endothelial cell-expressed S1P1 is now well-appreciated; S1p1-/- embryos die in utero at e12.5- E14.5 as a result of edema and hemorrhage, despite development of a vascular network 93. Close examination of S1p1-/- embryos revealed defective coverage of vessels by pericytes and vascular smooth muscle cell (VSMC) in several large vessels, including the dorsal aorta and the cerebral artery 93,94. Further studies with EC- or VSMC-specific deletion of S1P1 in vivo revealed that EC-intrinsic S1P1 is required for vessel coverage by VSMC 95. It is now known that S1P1 regulates both vessel maturation by regulating recruitment of mural cells needed for blood vessel stabilization, and the integrity of vessel barrier function by regulating adherens junctions (AJ).
5.1.1 S1PR in vascular barrier function and contractility
The signaling mechanisms involved in both cell migration and cell-cell junction formation are complex, involving the coordinated signaling of many synergistic and sometimes opposing pathways. Both junctional assembly and migratory responses are positively regulated by S1P1, and to some extent, S1P3, whereas destabilization and migration inhibition are mediated by S1P2 and possibly S1P3 signaling 96-98. Engagement of S1P1 activates solely Gαi, but the downstream signaling outcome can be cell-specific 99-102. Thus, S1P1 ligation can lead to increased intracellular Ca2+ mobilization and PI3K activation leading to Akt and/or ERK 1/2 phosphorylation, or Rac activation, subsequently inducing actin rearrangement, lamellapodia formation, and redistribution of junctional proteins, such as Zonula Occludens-1 (ZO-1), VE-cadherin, α- and β-catenin, and p-120 catenin to lipid rafts in the plasma membrane or the cell surface, culminating in the formation of endothelial AJ or TJ, or the migration of VSMC or EC. This can lead to decreased vascular permeability or increased vessel stabilization/angiogenesis 96,103-106.
The contribution of S1P3 to vascular barrier function has been more difficult to dissect, as it couples not only to Gi, but also to Gq and G12/1399. Mice deficient in S1P3 do not display any gross phenotypic abnormalities 107,108, and when S1p1-/-S1p3-/- embryos were generated, the bleeding phenotype was only slightly more severe than the S1p1-/-single mutant, likely indicating a supportive role for S1P3 in the maintenance of vascular barrier homeostasis and vascular development. Early in vitro studies demonstrated that S1P3-mediated RhoA activation was specifically required for the formation of stress fibers, in concert with S1P1-induced cortical actin assembly, and mouse embryonic fibroblasts (MEF) from S1p3-/- mice had defective activation of PLC and intracellular Ca2+ mobilization 96,107,108. Recent reports in other systems support this earlier work, showing that RhoA-mediated mechanical force and activated Rac are necessary for the formation and stability of AJ 109,110,111.
In vivo studies utilizing a model of acute stress, LPS administration, demonstrated that S1P3 /RhoA/ROCK activation dramatically increased vascular permeability 112. This was mediated by suppression of activated protein C (apC), a clotting regulatory factor that protects from endotoxin-induced lethality by countering the resultant thrombin-mediated vascular leak. Mice deficient in apC were more susceptible to LPS-induced vascular leak than wild-type mice, a phenotype that could be countered by the delivery of an S1P1 agonist or mimicked by S1P3 deficiency 113. The proposed mechanism for the barrier disruptive action is the stimulation of S1P3 activating RhoA, and its subsequent association with ROCK1, which is known to destabilize endothelial barrier function, and has also been demonstrated in vitro with LPS stimulation or TNFα administration to increase ROCK1 in lung EC 114,115.
Although results of these studies are seemingly at odds with work suggesting the cooperative effort of S1P1 and S1P3, it is not difficult to imagine a model taking into account the necessity of a dynamic barrier with the ability to withstand constant fluctuations in mechanical forces of homeostatic stress with relatively constant concentration of S1P present in the circulation. Under these conditions, S1P1 and S1P3 can function coordinately to allow the maintenance of barrier integrity while ensuring the passage of macromolecules into the interstitial compartment and the sampling of the blood by tissue-resident immune cells in the extravascular compartment. Under inflammatory conditions, possibly with increased S1P concentrations and altered concentrations of other inflammatory mediators in plasma, this balance could be manipulated. For example, splice variants of CD44, the hyaluronic acid receptor, differentially associated with and transactivated S1P1 or S1P3, depending upon the inflammatory environment 116. Consistent with this premise, whereas many studies show barrier-protective effects of S1P, high S1P concentrations can be barrier-disruptive in concert with LPS administration in vivo, an affect that can be attenuated by S1P3 knockdown or S1P2 knockout 117. Additionally, prolonged agonism of S1P1 has recently been shown to induce pulmonary vascular leakage and exacerbate lung injury in a bleomycin-induced injury model 118.
S1P2 appears to oppose many of the actions in the vasculature resulting from S1P1 signaling, due to the ability to couple with G12/13, initiating activation of the RHO/ROCK/PTEN signaling cascade 99,119,120. Although its biological functions are not as well-characterized, S1p2-/- mice have no obvious defects; however, they become deaf by one month of age due to defective vascularization of the stria vascularis, the tissue responsible for maintaining the endocochlear potential essential for audition 121,122. Evidence of the cooperative functions of S1P2 with S1P1 or S1P3 is found through studies using double- or triple-null mice. Whereas the S1P2 deficiency alone has little effect, S1p1-/-S1p2-/- embryos demonstrated earlier hemorrhage at E10.5 and dramatically increased perinatal lethality as compared to the S1p1-/- single gene deletion 94,108. S1p2-/-S1p3-/- double deletion also resulted in increased perinatal lethality compared to either single null, with few pups surviving to weaning age, presumably due to defective RhoA activation, as demonstrated using MEFs in vitro108.
Although S1p2-/- deficiency does not have the developmental effects of S1p1-/- deletion, the counter-regulation of S1P1 versus S1P2 has been well-illustrated using models of endothelial or vascular smooth muscle cell migration, vascular permeability, or vasoconstriction. In vitro, stimulation of EC S1P2/Rho/ROCK/PTEN disrupted AJ while stimulating stress fiber formation and increasing paracellular permeability 120. In vivo, H2O2-induced pulmonary vascular permeability was inhibited in mice treated with the S1P2 antagonist, JTE-013. Similarly, JTE-013 administration protected against S1P- and histamine-induced venular leakage in the cremaster muscle, in accordance with the enhancement of barrier function resulting from stimulation of S1P1 with FTY720, or the S1P1-specific agonist SEW2871123. Similarly, whereas S1P1 agonism or S1P2 deficiency protected against LPS-induced pulmonary vascular leak, S1P1 heterozygosity resulted in significant vascular barrier disruption, with or without exogenously administered LPS 117 However, another study challenged these conclusions, stating that platelet activating factor (PAF)-induced vascular permeability, which could be prevented by exposure to S1P, could only be reversed by S1P1 antagonism, and not antagonism of S1P2 or S1P3 124.
The discrepancies between these studies may suggest specific actions of different vascular stress stimuli. PAF receptor signaling occurs primarily via Gi, which could affect S1P1 signaling heterologously, whereas the histamine receptors may signal using several heterotrimeric G proteins 125,126. H2O2 stimulation induces not only PAF signaling, but VEGF and eNOS production and PLD activation, which could have diverse effects on S1P receptor signaling 127. Additionally, whereas S1P1/Gi activation can induce Akt and Rac-regulated eNOS phosphorylation and cell motility, S1P2/G12/13 or /Gq can inhibit chemotaxis by activating Rho 98,128. Thus, it would be expected that these different inducers of vascular permeability might engage signaling by different S1P receptors. In vivo, these would all be working in concert to determine the level of barrier permissivity. Additionally, vascular beds from different organs can exhibit differential responsiveness to S1P and other vasculature modifiers 129-131.
This is certainly true with regards to vascular contractility, which appears to vary depending upon the level of expression of S1P receptor subtypes. Activation of Gi/Akt induced eNOS-dependent vasorelaxation via agonism of S1P3 in isolated rat aortae or S1P1 in bovine lung EC 104,132. S1P3-deificent mice demonstrated significantly blunted vasodilatory responses after intraarterial administration of HDL or isolated sphingolipids 132. Conversely, coronary flow was decreased, and cerebral, mesenteric, or renal arteries were shown to contract ex vivo and in vivo in response to S1P signaling via S1P1 or S1P3, with S1P3 possibly antagonizing eNOS induction by S1P1 in some systems 131,133-136. Basilar arteries from S1p3-/- mice were refractory to S1P-induced contractile responses, whereas arteries from S1p2-/- mice responded with increased contractile sensitivity in response to S1P 131. Interestingly, this same study demonstrated that cerebral arteries, but not arteries isolated from the periphery, contracted in a dose-dependent manner in response to phospho-FTY720131. Another study demonstrated that lungs from wild-type mice treated ex vivo with JTE-013 and lungs from S1P2-deficicent mice demonstrated decreased S1P-induced pulmonary vasoconstriction, while treatment with the S1P3 antagonist VPC23019 had no effect 137. However, the specificity of both JTE-013 and VPC23019 has been called into question 131,138. These studies further highlight the differential responsiveness of vascular beds from various organ systems, as well as the complexities encountered when employing pharmacological tools.
5.1.2 S1PR regulation of angiogenesis
Cardiovascular and lymphatic angiogenesis are also regulated by S1P receptors, mediated primarily through regulation of cell motility, migration, and proliferative responses. Lymphangiogenesis may be positively regulated by S1P1/PLC-induced Ca2+ increases, as S1P1 RNA interference or S1P1 antagonism prevented S1P1-mediated lymphatic EC migration and proliferation in vitro 139. Many studies have documented the ability of S1P1 signaling to induce the migration of vascular EC and SMC, both of which are required for the development of new vessels and their stabilization 95,140-142. For example, in vitro antagonism of S1P1 inhibits vessel tube formation, whereas in vivo antagonism prevents angiogenesis in several model systems 142-145. Embryos with EC-specific S1P1 deletion display hemorrhage phenocopying that of the global S1p1-/- embryos 94,95. S1P2 may also play a role in the recruitment of cardiomyocyte progenitors: in zebrafish, the mutation of the S1P2 orthologue miles apart (mil) results in defective primitive heart tube formation evident as cardia bifida 146.
5.2 LPA receptors
The role of LPARs in the vasculature is not as well-defined, despite the fact that studies of ATX-deficient mice clearly demonstrated severely impaired embryonic vasculogenesis, and LPA induced decreased vascular barrier integrity via Rho/RhoA kinase signaling in HUVEC 18,42,147. Signaling via chicken orthologues of LPA1-3 in the chorio-allantoic membrane (CAM) assay demonstrated that LPA was angiogenic, most likely through LPA3 148. Although agonism of HUVEC LPA1 and LPA3 induced adhesion molecule and pro-inflammatory cytokine up-regulation, LPA1, LPA2, or LPA3 deficiency do not result in the dramatic lethality evident in S1p1-/- mice, thus originally making it difficult to determine the relevance of these receptors in the vascular system 149-152. No defects in blood or lymph vessel formation were grossly apparent in any of these mutant mice, although a small percentage of Lpa1-/- pups demonstrated frontal hematomas, a phenotype that was compounded in double deletants Lpa1-/-Lpa2-/- 150,151. Interestingly, zebrafish with LPA1 knocked down did not have abnormal blood vessel development, but rather defective embryonic lymphatic vasculogenesis 153. More recently, it was reported that almost 20% of Lpa4-/- embryos at E14.5 demonstrated increased perinatal lethality, with embryos displaying edema and hemorrhage at numerous anatomical sites 154. These phenotypes were the result of dilated blood vessels, defective lymphatic vessel development, and impaired mural cell recruitment. Interestingly, a previous study had shown that LPA induced VSMC migration and proliferation via receptor(s) that couple to Gq, although LPA4 had not yet been discovered and demonstrated to couple to Gq 155,156. This contrasts with findings from another group using Lpa4-/- MEFs, which demonstrated increased Akt/Rac signaling-mediated migratory potential 157; however, these cells were derived from mice with Lpa4-/- on a mixed genetic background, which did not display the same perinatal lethality of Lpa4-/- on a pure genetic background. How the genetic background affects vascular effects of LPA4, or indeed, any effects of the LP receptors, illustrates the need for cautious investigation.
Despite the lack of dramatic vascular defects in Lpa1-3-/- mice, studies have shown the importance of these receptors for maintenance of vascular homeostasis and modulation of inflammatory tone. Most recently, antagonism of LPA3 alone, or both LPA1 and LPA3, demonstrated that these receptors could positively regulate angiogenesis by down-regulating MVEC expression of CD36, the thrombospondin receptor and a potent anti-angiogenic signal 158. In other models of vascular dysfunction, such as neointima formation, expression of LPA1 and LPA2 by SMC modulates their migration to sites of vascular injury and neointimal lesions, whereas LPA3 in rat ASMC appears to mediate LPA-induced dedifferentiation 159,160. Additionally, bleomycin-induced pulmonary vascular leakage was dramatically reduced in Lpa1-/- mice 161.
LPA5 and LPA6 have only recently been described and determined to be bona fide LPA receptors, and thus far they do not appear to play any significant role in the regulation of endothelial cells 162-164. However, LPA5 was recently shown to regulate platelet activation, which can have dramatic effects on the cardiovascular system overall165.
6. Immune system
Many of the S1P and LPA receptors have been shown to play important roles in immune cell biology, especially with regard to the modulation of cell recruitment 30,166-169. In fact, some of the receptors are expressed with near exclusivity on cells or in organs of the immune system. Perhaps the most comprehensively explored role of an LP receptor in regulating immunity is the ability of S1P1 to regulate T cell migration into and out of the lymph node 167. However, LPAR and S1PR appear to play key roles in modulating the activity and migration of both innate and adaptive immune cells. Thus, in this review we will address the innate immune system first, as the first stage of the immune response.
6.1 Innate immunity
6.1.1 Neutrophils
Neutrophils are considered the first responders in most inflammatory responses. Because these granulocytes possess the ability to produce potent lipids, chemotactic factors, catabolic enzymes, and reactive oxygen and nitrogen species, their recruitment and activation are tightly controlled 170. Multiple studies have found LPAR and S1PR mRNA expression by human neutrophils, but the ability of LP to modulate neutrophils via GPCRs appears to depend upon their activation status171-173. Incubation with LPA or S1P in isolation is relatively ineffective for inducing changes in cell morphology, chemotaxis, or activation status 171,172. However, the response of neutrophils exposed to inflammatory or chemotactic stimuli, such as IFNγ or IL-8, is modulated by treatment with LPA or S1P through G protein signaling 173,174. Additionally, neutrophil expression of LPA1 and S1P3 were dramatically increased in neutrophils isolated from peripheral blood of patients with pneumonia, and were more responsive to chemotactic stimuli than those from healthy subjects 173. These differences are intriguing in light of the recent study demonstrating that neutral sphingomyelinase, a modulator upstream of S1P synthesis, polarizes the distribution of Rac1/2 versus RhoA at the leading edge of elongated neutrophils 175. Additional evidence of the effect of S1PR on neutrophil migration was demonstrated in studies using Sgpl-/- mice 176: S1P lyase deficiency increased neutrophil numbers in circulation by impairing homeostatic chemotaxis into tissues. As this phenotype was partially normalized by S1P4 deficiency, it is possible that S1P4 is responsible for regulating neutrophil migration during inflammatory responses that increase S1P concentrations.
6.1.2 Eosinophils
Eosinophils (EO) and mast cells (MC), granulocytes important for anti-parasite and anti-bacterial immunity, are thought to play important roles in the development and propagation of many inflammatory conditions where S1P or LPA are implicated, such as airway and skin inflammation 177-181. Although treatment with FTY720 or S1P1-specific antagonists can reduce airway inflammation, in some instances this can be attributed to reduced inflammatory status of the endothelium, and subsequently decreased chemokine production or inhibition of lymphocyte migration 182,183. In fact, a recent report states that administration of S1P can aggravate airway inflammation in a mouse model of allergen challenge 184. Thus, the many players of the inflammatory response and the models used can complicate understanding of LPAR and S1PR on EO and MC.
In IL-5 transgenic mice, the resultant eosinophilia can be reversed by treatment with S1P receptor modulators FTY720 or SEW2871 185. EO from these mice express S1P4 ≪ S1P1 < S1P3, and migrate toward S1P in vitro, a response that can be largely attenuated by pre-treatment of the mice with FTY720 or SEW2871. Additionally, human EO isolated from patients with allergic rhinitis also expressed S1P1 and S1P3-5, and this expression was up-regulated upon allergen challenge 186. S1P alone induces chemotaxis of human EO, but also induces up-regulation of other chemotaxis-related proteins, such as CCR3 and RANTES 187. Human EO also express LPA receptors, and it was reported that LPA induced migration, CD11b up-regulation, and induction of respiratory burst in vitro via LPA1 and LPA3 188. However, in studies using a collagen matrix, although LPA induced significant activation of RhoA, it was unable to induce EO migration through the 3D matrix 189.
6.1.3 Mast cells
The role of S1PR and LPAR in the regulation of mast cell biology has been extensively characterized. Both LPA and S1P have both been described as modulating mast cell activation state upon FcεRI ligation 190,191. Human MC express LPA1-5, and their development from monocyte precursors can be accelerated by addition of LPA to growth medium, increasing granule development and tryptase content, Kit expression, and proliferative capacity via LPA1 and/or LPA3 in a Gi-dependent manner 192,193. However, histamine release and chemokine production by human MC in response to LPA was IL-4-dependent, and regulated by LPA2 signaling 194. This was also true in rat and mouse MC, where LPA induced histamine release in a ROCK dependent-mechanism 195. Most recently, it was reported that LPA5 was the most abundantly expressed LPA receptor on human MC, and production of the chemokine MIP-1β was almost entirely due to LPA5 agonism 193.
S1PR appear to play a similar role in the activation of MC, including inducing the synthesis of the ligand via activation of sphingosine kinase, supporting an autocrine feedback loop 196. In vitro studies found that FcεRI ligation resulted in Sphk1 trasnlocation and production of S1P; however, a more recent study using Sphk1 and Sphk2 knockout cells and mice suggested that Sphk2 was necessary for intracellular S1P signaling, regulating FcεRI Ca2+-dependent effector responses, such as LTB4 and IL-13 release, whereas Sphk1-derived S1P from sources other than MC was necessary for S1PR activation of histamine release and the anaphylactic response 46,197. Exogenous S1P also induced the production of several CC chemokines, including MIP-1β, and induced cortical actin rearrangement and migration in bone marrow-derived mouse MC 197. In both human and rodent MC, S1P1 was able to induce cell migration, whereas S1P2 signaling regulates other activities, such as degranulation and chemokine release 197,198. Interestingly, whereas S1P2 was necessary for the induction of passive systemic anaphylaxis, induced by sensitization and subsequent challenge with IgE, it was subsequently found to be necessary for recovery from histamine-induced anaphylaxis, which obviates mast cell degranulation, leading to the hypothesis that S1P2 is necessary for histamine clearance, possibly via modulation of renal vascular tone 47,198.
6.1.4 Macrophages
Less is known about the role of LP receptors on macrophages, but much of what we understand of the actions of S1P and LPA in these cells have been determined through the use of autoimmune and infectious disease models or the use of the agonist/functional antagonist FTY720. Mouse macrophage expression of S1PR mRNA is dependant upon their origin; bone marrow macrophages express S1P1 and S1P2, whereas thioglycolate-elicited macrophages express S1P1-4, with S1P1 and S1P2 being expressed at equally high levels 199-201. Expression by alveolar macrophages from rat and mouse differed, with S1P2 and S1P4 being the only S1PR mRNA expressed by rat, but mouse alveolar macrophages expressed S1P1-4, with S1P2 predominating 202,203. Human peripheral blood monocytes (PBM) and PBM-derived macrophages produced varying levels of mRNA for S1P1-5 and LPA1-5 expression which could be altered dependent upon phenotype, i.e., differentiation to foam cells could increase or decrease expression of certain receptors, and human alveolar macrophages also exhibited a different receptor pattern 81,204,205. Differences in receptor expression can likely be explained by cell location and isolation methods, as monocytes and macrophages are notoriously susceptible to alterations in activation status and protein expression caused by manipulation 206,207.
S1P2 negatively affects macrophage migration in vitro in response to C5a complement component or CXCL12, as S1p2-/- macrophages did not display the increased cAMP or decreased Akt phosphorylation evident in WT cells 199. This is evident in models of atherosclerosis, where S1P2-/-ApoE-/- mice demonstrate reduced plaque formation and lipid deposition possibly due to an increased ability of macrophages to exit atherosclerotic vessels and/or regulation of the inflammatory phenotype, as S1p2-/- mice had decreased levels of serum pro-inflammatory mediators 200,208. Furthermore, S1P3 deficiency had no affect atherosclerosis plaques size, but significantly decreased macrophage recruitment to lesions and impaired chemotaxis and chemokine production in vitro; however, another study utilizing the NOD mouse model found that S1P3-/- monocytes were not defective in their ability to adhere to aortic endothelium ex vivo, and rather, S1P1 was responsible for this interaction, as demonstrated using the S1P1 agonist SEW2871 209,210.
Interestingly, apoptotic cell-derived S1P was shown to induce an anti-inflammatory “M2” phenotype in RAW264.7 macrophages via S1P2, as demonstrated by siRNA knockdown or JTE-013 antagonist 211. Primary human macrophages were also themselves protected from apoptosis, presumably via S1P1, as pre-incubation with phospho-FTY720 blocked this protection 212. In another study, however, S1P decreased LPS-induced pro-inflammatory cytokine production through S1P1 agonism, thus inducing an M2 phenotype through this receptor 201. Some of these differences might be due in part to the inflammatory stimulus, and have implications for the mechanism of continued monocyte/macrophage activation in atherosclerosis and chronic inflammatory disease, as the number of necrotic versus apoptotic cells increases with the severity of atherosclerotic disease 213. S1P2 also appears to modulate the macrophage response to infectious agents, as S1p2-/- alveolar macrophages expressed reduced levels of Fc receptors, and were compromised in their ability to phagocytose the fungal pathogen Cryptococcus neoformans 203.
Although LPA has long been known to act as a macrophage survival factor, it also modulates the activity of mouse and human macrophages 202. Through a Gi-dependent pathway, LPA up-regulated ROS production in mouse and human alveolar macrophages as well as Ca2+ mobilization, IL-1β and TNFα transcription and protein expression in the mouse J774A.1 macrophage cell line and human monocyte-derived macrophages 81,202,204,214. Cell migration was also controlled by LPA1 and/or LPA3, as monocytic cells appeared to migrate toward concentrations of LPA < 1 μM, whereas migration is inhibited above this concentration in vitro 215. The potential relevance of these receptors to the modulation of monocyte and macrophage recruitment during disease states is evident in models of atherosclerosis, where it was recently shown that macrophages in human atherosclerotic plaques expressed both of these receptors 216. Thus, LPA participates in a neatly organized amplification feedback loop: with LPAR expressed by both endothelial cells and monocytes/macrophages, LPA can activate both cell types, activating endothelial cells to produce myeloid recruiting chemokines, as well as LPA itself. These chemokines then coordinately recruit myeloid cells, which along with LPA, can also activate these cells to produce pro-inflammatory mediators, such as IL-1β and TNFα, further activating the nearby endothelium, and amplifying the response 149,216,217.
6.1.5 Dendritic cells
Despite evidence that large amounts of both S1P and LPA are produced during some immune and inflammatory responses, the role of LPA in dendritic cells has not been widely explored. Immature dendritic cells (DC) derived from human peripheral blood monocytes express mRNA for LPA1-5, and this expression is modulated upon maturation, with LPA3 becoming significantly down-regulated 218-220. Accordingly, LPA differentially affects the activity of these different sets of DC: in immature DC, unsaturated LPA induces intracellular Ca2+ mobilization, actin polymerization, and chemotaxis 218,220. When added to maturation medium, those DCs matured in the presence of LPA induced a Th2 phenotype in T cells, and augmented their proliferative capacity in vitro 218,219. Thus, LPA may play an important role in determining the tone of the nascent acquired immune response, but more study is needed to see how it specifically affects DC biology.
Many of the activities of S1P on DCs are reminiscent of those of LPA, with important roles in migration and phenotype. In fact, S1P4 was cloned from mature human DC 221. Human and mouse immature and mature DC express mRNA and protein for S1P1-5 222-224. Similar to LPA, S1P induces intracellular Ca2+ mobilization, actin polymerization, and migration in immature DC, and appears to induce a shift from Th1 to Th2 phenotype in DC that are matured in the presence of LPS 222. Perhaps one of the more intriguing differences is the ability of S1P to induce the migration of DCs that are matured with TNFα and PGE2 versus those matured with LPS, and DC that originate from the peripheral blood, bone marrow, or skin 222,224-226. For instance, the migration of LPS-matured DC appears to be dependent upon S1P3 and not S1P1, as demonstrated using cells from S1P3 knockout mice, but TNFα/PGE2-matured DC appeared dependent upon S1P1; however, the mRNA of both receptors is up-regulated upon stimulation 223,225,226. In vivo studies using FITC-dextran recruitment or adoptive transfer of S1p1-/- or S1p3-/- DC illustrated the differential dependence of in vitro matured DC versus mature skin DC on both S1P1/3 or S1P1 alone, respectively, in their migration to lymph nodes 226.
Tissue-resident DC, such as Langherhans cells (LC) or lamina propria DCs, are phenotypically different from other DC, and as such might be expected to be differentially affected by S1P 227. In fact, whereas SEW2871 prevented LC migration to draining LN, implying that LC require only S1P1 for migration, lamina propria DC appear to require both S1P1 and S1P3 to exit the gut and migrate into mesenteric LN, as S1P3-/- mice have impaired DC migration from LP in response to LPS stimulation, but LC migration is unaltered 225,226. A separate study stated that skin DC were impaired in their ability to chemotax in response to various chemokines in the presence of the S1P1-specific agonist SEW2871 224. Interestingly, S1P1-mediated LN migration is negatively regulated by CD69, which performs the same role in T cells, a regulatory mechanism specific for S1P1 228,229.
S1P mediates other aspects of DC biology besides migration, although it does not appear to be necessary for maturation 226. LC that had been exposed to FITC-dextran and migrated to LN expressed higher levels of mRNA for the S1P1 and S1P3 than did LC still resident in the skin 225. Although partially reflective of LC dependence on the S1PR for migration, it was also reported that S1P3 was necessary for endocytosis, a step that would naturally occur before these cells traveled to the LN for antigen presentation to T cells: phospho-FTY720 impaired FITC-dextran endocytosis, whereas SEW2871 did not 223. Studies using S1P3-deficient mice demonstrated that S1P3-regulated IL-1β production by DC is also a critical regulator of systemic inflammation induced by sepsis, illustrating that S1P3 signaling likely plays a role in modulating the production of various DC-derived cytokines 113,222.
6.1.6 Natural killer cells
Natural killer (NK) cells are innate immune cells important for killing transformed cells, primary defense against infectious organisms, especially viruses, and directing the acquired immune response through the production of interferons and other cytokines and chemokines 230. Not much is known about how these cells are regulated by LPAR and S1PR, although human and mice NK cells do express the mRNA for LPA1-3, and S1P1-5 231-233. As in many other immune cells, LPA induces intracellular Ca2+ mobilization, F-actin polymerization, and migration in a concentration- and Gi-dependent manner 231,234. Curiously, although LPA increases NK cell IFNγ production, it decreases perforin production and cytotoxic activity through LPA2 agonism and subsequent cAMP/PKA activation 231,234.
Although NK cells appear to express all of the known S1P receptors, the receptor known to have any major role in the regulation of NK activity is S1P5 235. S1P5 expression in NK is regulated by T-bet, a transcription factor critical for the decision between CD4 and CD8 cell fate 236,237. NK cells migrate in response to S1P, and the expression of S1P5 regulates bone marrow and lymph node egress of NK cells 232,233,237. Other roles for S1P5 or the other S1PR have not yet been reported.
6.2 Acquired immunity
6.2.1 Natural killer T cells
Natural killer T cells are a cellular bridge between the innate and acquired immune systems. They are thymocytes; however, they express the NK receptor and are characterized by a restricted T cell receptor (TCR) which recognizes only glycolipid antigens presented on the MHCI-like CD1d 238. Little is known about how the expression of LP receptors might affect these cells; indeed, it is unknown whether they express any of the LPAR, although pulsing antigen-presenting cells with LPC induced increased cytokine production by NKT clones and primary polyclonal human peripheral blood NKT cells 239. Although it has been demonstrated that NKT express S1P1, 2, & 4 mRNA, S1P1 is thus far the only receptor whose role has been examined 240. Using inducible S1P1 knockout mice, it was demonstrated that S1P1 is not necessary for NKT precursor development and migration to the thymus, but is required for their normal distribution throughout the tissues 240. Although a subsequent paper reported that FTY720 treatment did not impair NKT migration but did alter cytokine responses, these responses occurred in a Gi-dependent manner, indicating that other S1PR, including S1P5, could be involved 241. It is curious that, despite the important role of S1P5 in regulating NK migration, this receptor has not yet been examined in NKT cells.
6.2.2 T cells
A great deal is known about the role of S1PR in the regulation of T cell responses. The predominant S1P receptors on T cells appear to S1P1 and S1P4, although mRNA for the other receptors can be found, albeit at low levels 242. T cell surface expression of S1P1 is perhaps the major determinant of T lymphocyte egress from the thymus and secondary lymphoid organs, overriding the retention signals transduced by CCR7 signaling, and varies in a cyclical manner, inversely correlating with the S1P concentration gradient 19,242-248. Whereas single-positive (SP) CD4+ or CD8+ T cell S1P1 expression is high in the thymus, it is low in the blood, up-regulated once again upon entry into the LN, and then decreases once again upon exit into the lymph 245. This cyclical expression is orchestrated, in part, by CD69 expression, which is up-regulated immediately upon entry into the LN, and negatively regulates S1P1 through direct interaction with the receptor, facilitating the retention of cells within the lymphoid organs 229,249,250. Forced expression of S1P1 on double positive (DP) and immature SP cells, which under normal circumstances is directly controlled by the transcription factor Kruppel-like factor (KLF)-2, is sufficient to induce premature egress from the thymus 251,252. Specifically in differentiated Th2 cells within the LN, extracellular matrix protein-1 (ECM1) drives S1P1 re-expression after antigen education via KLF-2 induction, permitting LN egress 253. Additionally, expression of S1P1 modulates the ability of effector T cells to migrate out of inflamed tissues into afferent lymphatics, and is a factor in the decreased chemotactic capacity of T cells from old versus young human subjects 254-256.
S1P1 expression is also important for T cell maneuvering within the LN and tissue-specific homing. Entry into LYVE1+ cortical sinuses is also regulated by S1P1 expression, as S1P1-deficient cells display sinus probing behavior similar to that of WT cells, but their entry into sinuses was ≤ 20% that of WT controls 257. S1P1 signals determine the migration of conventional SP T cells to become intraepithelial residents of the intestine, and T cell rolling and sticking in peripheral LN high endothelial venules (HEV) was reduced in S1P1-/- mice, but was unaffected in the Peyer's patches (PP) 258,259. Interestingly, Halin, et al. also indicated the potential involvement of other S1PR in the modulation of PP T cell trafficking, as treatment with FTY720 enhanced PP T cell retention in both WT and S1P1-/- mice.
S1P1 can also modulate T cell subsets, which is especially evident in S1P1 T cell-specific Tg mice 260. Transduction of S1P1 into a non-expressing T cell line lead to S1P-mediated reductions in IFNγ and IL-4 production by murine splenic CD4+ cells and inhibition of proliferative responses to anti-CD3/CD28 stimulation 261. In mice bearing both the OTII OVA-specific TCR Tg and the T cell-specific S1P1 Tg, the Th17 response was enhanced, with increased numbers of Th17 cells producing greater concentrations of IL-17 as compared to single Tg OTII OVA mice 262. Similarly, WT T cell development of the Th17 lineage in vitro was induced by S1P to the same extent as that induced by IL-23, and this differentiation could be recapitulated by SEW2871, or prevented by phospho-FTY720, indicating that S1P1 may be an important modulator of the Th17 inflammatory response 263.
T regulatory (Treg) cell development also appears to be dependent upon S1P1 signaling. Administration of FTY720 increased the number and suppressive activity of splenic CD4+CD25+Foxp3+ Tregs, which express lower levels of S1P1 and S1P4 mRNA, and higher levels of S1P3 mRNA than do CD4+ effector cells 264,265. Recent studies have demonstrated that S1P1 blocks the thymic generation of CD4+CD25+Foxp3+ Tregs by activating the AKT-mTOR pathway, preventing the differentiation of CD4+CD25+Foxp3-precursor cells 266. Sustained stimulation of mTOR and suppression of TGFβ-Smad3 signaling by S1P1 in peripheral T cells leads to the preferential generation of Th1 effectors over induced CD4+CD25+Foxp3+ Tregs 267. Although both conventional SP cells and Tregs lose expression of S1P1 upon TCR complex engagement, Tregs appear to down-regulate S1P1 more slowly 266. This elegant regulatory system prevents immune suppression from occurring too early during the immune response, but leads to its eventual termination.
LPAR also have a role in regulating T cell responses, although it is not as well understood as that of S1PR. Human CD4+ and CD8+ SP express predominantly LPA1 and LPA2, and low levels of LPA3 268,269. LPA suppressed Bax-mediated apoptosis in a human T lymphoblastoma line, inhibited activation-induced CD4+ IL-2 production, and augmented IL-13 production via LPA2 in human peripheral blood T cells 180,268,270,271. LPA2 also stimulated CD4+ cell migration, but LPA1 blocked migration of Jurkat T cells toward RANTES (CCL5) 272. The LPA5 receptor is the most highly expressed LPAR in CD8+ cells of the gut epithelium; however, no further studies have been reported with regards to LPAR in this group of cells 162. The effects of LPAR on T cell immunity are likely to be highly significant, as ATX is expressed on HEV of LN, and administration of an enzymatically inactive form of ATX impaired lymphocyte homing to secondary lymphoid organs (SLO) 90.
6.2.3 B cells
The role of S1PR and LPAR in the regulation of B cell responses has not been explored as thoroughly as in T cells. In fact, very little is known about the role of LPAR in normal B cells, as most research has been conducted using B cell cancer lines. LPA1 is up-regulated in and protects from apoptosis chronic lymphocytic leukemia (CLL) cell lines or primary patient CLL 273. LPA1, 3, & 4 are expressed in a human B cell line, which also expressed functional ATX, and mouse primary splenic and bone marrow B cells mobilize Ca2+ in response to LPA, although it is unknown which receptor this occurs through 274,275.
More information is available regarding the role of S1PR in B cells. Expression of both S1P1 and S1P4 increases with maturation stage, whereas S1P3 is most highly expressed in immature B cells 276,277. These changes in S1PR subtype expression may underlie the differential responsiveness of B cells at various stages of maturation. S1P1 is involved in the migration of immature B cells into the bone marrow vascular compartment and subsequently into the peripheral blood 21,276. B cell-specific knockout of S1p1 resulted in fewer immature B cells in the circulation and spleen without a concomitant increase in the number of immature cells present in the bone marrow parenchyma, although S1p1-/- B cells were less efficient at egress from the bone marrow 276,277. This lack of reciprocity was evident in S1P1-deficicent mice, as well as WT mice treated with FTY720, and was explained by increased apoptosis of immature B cells that could contact the bone marrow vasculature, but not enter 276,277. Interestingly, S1p1-/- immature B cells chemotax toward S1P, whereas S1p3 deletion rendered immature B cells unresponsive to S1P in vitro, and S1p3-/- mice had significantly fewer circulating immature B cells 276, Additionally, S1P3 expression by marginal zone (MZ) MAdCAM-1+ endothelial cells was required for their organization around B cell follicles, whereas migration and localization of the B cells themselves necessitated S1P1 expression 278,279. Curiously, despite several studies demonstrating reduced migratory capacity of S1p3-/- bone marrow and MZ B cells in chemotaxis assays, expression of this receptor has not yet been shown to have an intrinsic, in vivo effect on B cells 276,278,279.
Upon differentiation of mature activated B cells to IgG plasma cells, or antibody-secreting cells (ASC), differential S1P1 expression is regulated by KLF2 280. S1P1 then determines whether a specific ASC will be retained in the SLO, or migrate into other organs, such as the BM or possibly the mucosa, an effect seen with both S1p1-/- mixed bone marrow chimeras and FTY720 treatment 280. With regard to the intestinal mucosa, studies using FTY720 treatment demonstrate that S1P regulates the trafficking of peritoneal B cells and Peyers' Patch plasmablasts (immature plasma cells) into the intestinal lamina propria, thus regulating IgA production 281,282. Although no receptor-specific agonists, antagonists or genetically-modified mice were used in the studies examining ASC migration in the gut mucosa, the above-referenced study by Kabashima, et al. imply that S1P1 will likely play a role in the retention of these cells.
7. Stem cells
Hematopoietic stem and progenitor cells (HPC) have been shown to constitutively migrate through the blood, BM, peripheral tissue, and lymph 283. During an inflammatory response to zymosan or mobilization induced by G-CSF, S1P levels in peripheral blood increased, due in part to activation of the complement cascade, mobilizing HPC from the bone marrow 284. Other reports demonstrated that S1P-induced homing of CD34+ HPC was partially regulated by S1P3-mediated modulation of the CXCR4-JAK/STAT pathway, leading to increased SDF-1 responsiveness 285,286. Mononuclear cells from S1P3-deficicent mice were unable to home to sites of ischemic injury, and FTY720 pre-treatment increased homing of transferred CD34+ HPC injected via the tail vein or peritoneum to the bone marrow and sites of ischemic injury 285,286. Administration of FTY720 could improve neovascularization by patient-derived endothelial progenitors, as well as support proliferation and homing of HPC by drawing them out of the out of the lymph and blood into extramedullary tissues 283,285,286.
S1PR are also specifically involved in the migration of osteoclast precursors. SEW2871 treatment stimulated migration of osteoclast precursors from bone marrow into the blood, and mice lacking S1P1 expression by osteoclast precursors demonstrated increased osteoporosis and osteoclast attachment to bone surfaces 287. Additionally, FTY720 administration ameliorated bone loss in a mouse model of post-menopausal osteoporosis 287. Conversely, JTE-013 blocked S1P2 opposition S1P1-mediated chemotaxis of RAW264.7 cells (used as a model of osteoclast precursors) at high S1P concentrations 288. In vivo, JTE-013 administration largely reversed bone density loss induced by RANK ligand administration, an effect that was absent in S1p2-/- mice 288. Thus, osteoclast precursor mobilization in the marrow spaces appears to be reciprocally regulated by S1P1 and S1P2, as S1P1 attracts osteoclast precursors to the blood and S1P2 serves to repulse precursors from blood and maintain them in the bone marrow 287,288.
Early studies demonstrated that LPA1 expression correlated with HPC ability to invade stromal cell layers in vitro 289. Interestingly, LPAR also have a role in regulating bone morphogenesis, but do not appear to regulate migration. Rather, the LPA1/3 antagonist Ki16425 inhibited human mesenchymal stem cell (MSC) differentiation to osteoblasts, and as these cells did not express LPA3 mRNA, this was attributed to LPA1, whereas shRNA against LPA4 allowed for osteogenesis, indicating a negative role for this receptor 290. This was evident in Lpa4-/- mice, which had increased bone volume and density 290. Additionally, mice lacking LPA1 exhibited skeletal abnormalities and decreased bone content, due to decreased osteoblast differentiation from precursors 291. Additional roles for LPAR and S1PR in SC differentiation have been proposed, based upon data from nervous system and will be discussed in that context.
8. Nervous system
Of the LP receptors, LPAR predominate in regulation of nervous system biology. LPA1 was cloned from a neuronal cell line derived from the ventricular zone (VZ), and in the mouse embryonic neocortical VZ displayed enriched expression correlating with the stage of neuronal development, and was therefore originally named ventricular zone gene-1 (vzg-1) 11. Since that time, expression of LPA receptors by most cell types of the nervous system has been reported, with pleiotropic effects on the different neuronal cell types, including their development, migration, and signaling 63,292.
8.1 Neural progenitors
Neural progenitor cells (NPC) express high levels of LPA1, LPA2, and LPA4 mRNA, and therefore are capable of transducing diverse signals 63. The embryonic telencephalon, as a whole, expresses LPA1-5 mRNA 293. LPA induces neurite retraction, nuclear rounding, and positioning of VZ neuroblast nuclei to the apical surface in vivo, and stimulates NPC survival in vitro through LPA1/Rho activation294-297. Interestingly, the ability of LPA1 and LPA2 to coordinately inhibit VZ NPC cell death in the cerebral cortex are illustrated by the enhanced cortical growth and folding which results from exogenous LPA administration to cerebral explants, and the lack of folding in cortices of Lpa1-/-Lpa2-/- mice 298. NPC fate decision between neuronal and oligodendroglial is also determined by LPA, with LPA1/Gαi signaling leading to induction of neuronal differentiation while suppressing glial differentiation, as demonstrated using pertussis toxin or Ki16425 299. Many of these responses are dependent in part or in whole, upon Ca2+ mobilization or Ca2+ influx, and both LPA1 and LPA2 have been shown to evoke Ca2+ responses in NPC in vitro 293. Stable expression of LPA4 allowed for LPA-mediated, Ca2+-dependent morphological changes, including cell rounding, neurite retraction, aggregation, and N-cadherin-mediated cell-cell adhesion via the Rho/ROCK pathway 300. In differentiated mouse neuroblastoma cells intrinsically expressing or with stable transfection of LPA1, neurite retraction was mediated by LPA1 induction of PLC/Pyk2/GSK-3β or PKA signaling pathways 301,302.
S1P also plays an important role in the developing nervous system and NPC recruitment, as S1p1-/- embryos demonstrate significantly increased apoptosis and decreased mitosis in the neuroepithelial layers of the developing telencephalon 45. NPC expressed mRNA for S1P1-5 and respond to S1P by induction of Ca2+ mobilization similar to LPA stimulation 293,303. NPC also responded to S1P by forming aggregates and undergoing proliferation and differentiation, which could be prevented by PTX or inhibition of ROCK in vitro 303. In vivo, shRNAi silencing of S1P1 prevented the migration of transferred NPC to sites of spinal cord injury 304. Because of their known function in the regulation of hematopoietic stem cells, future studies are likely to uncover more functions of the S1PR in the developing nervous system.
8.2 Astrocytes
Astrocytes are cells that contribute to information processing and regulation of neural activity by networking with neurons and regulating brain metabolism 305. These cells express LPA1-5, as well as S1P1, S1P3-5 306,307. Although some debate exists as to the ability of LP to induce proliferation in vitro, in vivo injection of either LPA or S1P leads to astrogliosis 307. Induction of proliferation in vitro occurs through activation of Gi-coupled pathways, and both LPA and S1P can induce Ca2+ mobilization 307-309. Incubation with LPA mediated DNA synthesis and astrocyte proliferation while inhibiting their ability to uptake glutamate; the former was mediated by LPA1, as demonstrated by LPA1-/- cells; however, the LPAR responsible for the regulation of glutamate uptake was not identified 306,310. Coordinated LPA1 and LPA2 signaling in astrocytes also induced the production of a factor that encouraged neuronal differentiation, as medium from WT astrocytes primed with LPA induced astrocyte differentiation, whereas the medium from similarly primed, Lpa1-/-Lpa2-/- astrocytes did not 311.
Information regarding the role of S1P in these cells is not abundant; however it is known that S1P signaling via Rho/ROCK inhibits primary striatal astrocyte gap junction formation in vitro 312. Additionally, a recent study utilizing CNS cell lineage-specific deletion of S1P1 reported that non-immune mediated efficacy of FTY720 in models of EAE could be mediated through astrocyte-expressed S1P1 313. Further studies will likely be aimed at determining the precise mechanism by which FTY720 modulates astrocyte function and autoimmune inflammation in the CNS.
8.3 Microglia
Another glial cell type, microglia, are the tissue resident macrophages derived from primitive myeloid precursor cells, and are likely important for normal brain development 314. The level of expression of the LPAR varies between species; human microglial cell lines are able to express LPA1-3 315,316. LPS stimulation of rat microglia increased production of LPA3 mRNA, implicating a role for this receptor in inflammatory responses 317. LPA administration to microglia in vitro can induce Ca2+ signaling, proliferative responses, ATP release, ERK signaling, and membrane ruffling 315,317-319.
Microglia also express S1P1-3, 5 mRNA, and upon stimulation with LPS or agonism with S1P, S1P1 and S1P3 mRNA are decreased 317. S1P dose-dependently induced TNFα release from rat microglia, activated p38 signaling, and also altered expression of the mRNA for LPAR, demonstrating the interconnectedness of these two signaling pathways 317. Other studies on the role of S1PR in microglia have specifically examined the effects of FTY720. Although FTY720 induced down-regulation of phospho-ERK with a concomitant increase in phospho-MLC II in DC and macrophages, microglia p-ERK and p-MLC II were unaffected 319. FTY720 also increased the numbers of microglia in an ex vivo model of demyelination while decreasing their phagocytic capacity via S1P1 and S1P5, as determined by the use of SEW2871 and a Novartis S1P5-specific agonist 320.
8.4 Oligodendrocytes and Schwann cells
Oligodendrocytes and Schwann cells perform the task of myelination in different branches of the nervous system, CNS and PNS, respectively 321. The myelin sheath is composed approximately 70:30 of lipid, and LPA1 expression correlates with oligodendrocyte maturation stage, and subsequently, myelination status 322. Differentiated oligodendrocytes express LPA1, LPA3 and very low levels of LPA2, and LPA-initiated Ca2+ signaling and p-ERK are induced by MAPK/PLC signaling in a PTX-insensitive manner 323,324. LPA was also found to increase myelin basic protein (MPB) mRNA and the production of myelin sheets in vitro, although evidence for a direct effect on myelination by oligodendrocytes in vivo is lacking 324.
The first description of S1P5 stated that it was highly expressed in the white matter of the brain, implying expression in oligodendrocytes 325. Subsequently, studies utilizing cells from S1p5-/- mice demonstrated that S1P-mediated induction of process retraction and inhibition via Rho/ROCK of immature oligodendrocyte migration, and promotion of mature oligodendrocyte survival via Gi/AKT signaling occurred via S1P5 signaling 326,327. Administration of FTY720 increased proliferation of mature oligodendrocytes and induced phosphorylation of AKT and ERK via S1P1 signaling, whereas it promoted the maturation of oligodendrocyte precursors, which express high levels of S1P5 and low levels of S1P1, indicating that S1P5 might play a role in the beneficial effects of FTY720 by promoting the oligodendrocyte differentiation 328.
Although it does not affect oligodendrocyte survival, LPA is a critical Schwann cell survival factor, signaling via LPA1/Gi/PI3K/AKT 329. LPA1/Rho/ROCK signaling also mediates Schwann cell actin cytoskeletal changes, induces focal adhesion assembly, and N-cadherin-based cell-cell adhesion 330,331. The role of LPA1 in vivo was confirmed in Lpa1-/- mice, which demonstrated increased apoptosis of sciatic nerve Schwann cells 150. S1P, however, has not been shown to play a prominent role in Schwann cell biology, despite their expression of S1P1 and possibly S1P2 329. S1P also induced major morphological changes due to actin cytoskeletal rearrangement, although this was reportedly due to activation of Rac1 331.
8.5 Neurons
Many of the neuronal effects of LPA, such as growth cone turning, neurite retraction, and chemorepulsion maybe be mediated by receptors other than LPA1, as cortical neurons from Lpa1-/- mice were responsive to LPA 40,302,332. LPA3 is expressed on motor neurons of the spinal cord and the cortex and is up-regulated upon spinal or brain injury 333. LPA1 has, however, been shown to play an important role in nociception. Studies in Lpa-/- mice demonstrated that the LPA1/Rho/ROCK pathway mediated allodynia and hyperalgesia induced by LPA, as well as demyelination and neuropathic pain initiated by substance P production, sciatic nerve injury or compression of the trigeminal ganglion 334-337. Interestingly, LPA1 is involved in behavioral alterations unrelated to pain, including the suckling impulse of neonates, possibly regulated by LPA1-mediated development of the cerebral cortex or olfactory bulb, and regulation of exploratory behavior, spatial learning, and working memory 150,338.
There have been few reports claiming an important role for specific S1PR signaling in adult neurons. Dorsal root ganglion and sensory neurons express mRNA for S1P1-4, and S1P augments action potential of specific neurons by suppressing outward potassium current 339. In another study, in vitro agonism of S1P1 with SEW2871 increased the excitability of roughly half of the rat adult sensory neurons examined, also implicating other S1PR in the determination of neuronal sensitization 340. In Xenopus retinal ganglion cells (RGC), anti-S1P5 blocking antibody studies illustrated that the S1P5/RhoA/Lim kinase signaling cascade mediated growth cone chemorepulsion to S1P, and ex vivo S1P was an axonal guidance cue in retinal explant cultures 341. S1P also elicited a cation current mediated by PLC activation and iCa2+ in chicken Amacrine cells (retinal interneurons), possibly via both S1P1 and S1P3, as treatment with SEW2871 induced current, whereas suramin, a S1P3 antagonist, reversibly blocked current induction 342. However, caution is required when examining data obtained using suramin, as numerous other, non-S1PR-mediated effects have been reported 343,344.
9. Reproductive system
LPAR and S1PR have important roles in the process of reproduction under normal and disease conditions, in the parents as well as the offspring. Although early reports describe fluctuations in the concentrations of LPA, LPC, and S1P in the reproductive organs reflecting reproduction receptivity and capability, there is not much known about the contribution of specific receptors 345.
9.1 Female reproductive biology and gestation
Embryo implantation in the endometrium is a critical milestone in the pregnancy timeline and can be a primary contributor to infertility 346. Delayed embryonic development and increased embryonic death due to abnormal implantation and spacing resulted from maternal LPA3 deletion, consequently down-regulating COX-2 152. Pregnant Lpa3-/- mice administered exogenous PGE2 and PGI2, COX-2 products required for implantation and decidualization, demonstrated increased successful embryo implantation 152,346. Studies in pigs indicated that LPA3 expression may be spatially and temporally regulated, dependent upon the region of the uterus and phase of the estrus cycle 347. Interestingly, although LPA3 is highly expressed in oviduct, placenta, and uterus, it is absent from the ovary, where LPA4 is highly expressed in both human and mouse tissues 152,348. In humans, endometrial stroma and first trimester trophoblasts expressed only LPA1 mRNA, and LPA signaling induced in vitro IL-8 production, angiogenesis, and immune cell migration 349,350. These studies demonstrate that both maternal and fetal LPAR signaling cooperate to ensure successful pregnancy.
Signaling via S1PR also plays an important role in pregnancy. S1P1 was up-regulated in sheep endometrium and localized to areas of angiogenesis 351. S1P1, 2, & 3 mRNA were up-regulated in murine decidua and in uterine stromal cells, and immunohistochemistry demonstrated that S1P1 and S1P2 colocalized with COX-2 at various points of the maternal-fetal interface, indicating that S1P signaling may participate with LPA3 in regulating COX-2 expression in the uterus 352. However, a subsequent paper reported early pregnancy loss in Sphk1-/-Sphk2+/- mice due to defective decidualization, but no differences in uterine COX-2 expression nor PGE2 were detected 353. During late stages of pregnancy, S1P1, S1P2, and S1P3 were also detected in human decidua, with S1P3 expression altered according to gestational age 354. Additionally, S1P metabolism increased with gestational age, with highest decidual activity at term, which might prepare the decidum for parturition 351. This is an interesting notion, as COX products are critical for the induction of labor, and therefore S1PR may be important for inducing COX regulation of later stages of pregnancy 346.
9.2 Male reproductive biology
The male reproductive system is also regulated by LP receptors. High mRNA expression of LPA1-3 and low expression of LPA4 and LPA5 were detected in mouse testis, as well as the germ cells themselves 355. Single knockout males demonstrated a small decrease in sperm count at 6 months of age, and double knockout further decreased sperm numbers. LPA1/2/3 triple knockout males exhibited numerous reproductive deficiencies, including decreased mating activity, age-related testicular degeneration and decreased sperm counts related to increased germ cell apoptosis 355.
S1P is also a regulator of the male reproductive organs, although little is known about the role of specific receptors. S1P inhibits male germ cell apoptosis, and Sphk1 is expressed in the acrosomes of spermatozoa 356,357. Mature spermatozoa also express mRNA for S1P1-3 and S1P5, and S1P1 is highly expressed in seminiferous tubules and spermatids in the testis 358. S1PR may also play a role in male sexual function, by virtue of eNOS phosphorylation. Human corpus cavernosum expresses S1P1-3, and S1P signaling potentiates acetylcholine-induced eNOS activation, which in turn regulates the hemodynamics required for erectile function 359. Thus, S1PR and LPAR regulate the development and maintenance of both male and female fertility and reproduction.
10. Inhibitors, agonists, and antagonists
Development of S1PR- and LPAR-modifying compounds with in vivo utility has been hindered by numerous challenges, such as the lack of ligand-bound GPCR crystal structures, carrier/chaperone affinities, off-target effects, and conformational changes 360,361. Despite these difficulties, these receptors remain attractive targets, due to their disease state correlation, potent functional effects in numerous organs/systems, and prior amenability to modulation by pharmacological compounds. There are numerous agonists and antagonists of LPAR and S1PR, a few of which will be discussed herein according to receptor specificity/preference.
10.1 ATX inhibitors
Because of the proposed ability of autotaxin to deliver LPA directly to LPAR, some of the newest pharmacological tools, the autotaxin inhibitors, must be addressed in the discussion about LP receptor-modulating drugs. Previously, studies of ATX/LPA had been hindered by the lack of ATX inhibitors with in vivo potency 362,363. PF-8380 was obtained from a screen of the Pfizer compound collection and subsequent structure-function studies. Maximal inhibition of LPA synthesis in plasma occurred 15-30 minutes after administration, and a 30 mg/kg dose in rats showed efficacy equal to the same dose of naproxen in a model of adjuvant-induced arthritis 364. Small molecule screening and derivative optimization resulted in the synthesis of HA130, a boronic acid-based inhibitor targeted to the T210 active site of ATX. This compound, administered IV at 1 mol/g resulted in approximately a 75% decrease in plasma LPA within 10 minutes; however, no in vivo functional assays were performed in this report 365.
The most recent advance in autotaxin inhibitor development was the design of stable analogues (compounds 22 and 30b) of S32826 [4-(tetradecanoylamino) benzyl] phosphonic acid, a molecule that demonstrated an IC50 in the nanamolar range in a cell-free system, but poor stability precluded in vivo analyses 366,367. Both compounds 22 and 30b were highly specific for ATX, exhibited long plasma half-lives of approximately 10.5 hours, inhibited ATX-dependent invasion of HUVEC monolayers by rat hepatoma in vitro, and decreased metastatic foci in the B16-F10 melanoma model 367. The authors suggest that continuing compound evolution originating from the same benzyl- and naphthylphosphonic acid scaffold may yield ATX inhibitors with extended half-lives, and thus greater utility in the tumor microenvironment, characteristics which would likely prove useful in other disease models.
10.2 LPAR agonists and antagonists
10.2.1 LPAR antagonists
AM095 is a LPA1-specific antagonist from with efficacy in mouse models of fibrosis, scleroderma and bleomycin-induced pulmonary fibrosis without effects on wound healing 368,369. This compound led to the development of AM152, a LPA1 antagonist that has recently completed Phase I clinical trials for the treatment of systemic sclerosis (http://www.amirapharm.com/articles/AmiraAM152Phase1.html), demonstrated efficacy in numerous mouse models of fibrosis and was granted Orphan Drug Status by the FDA for the treatment of idiopathic pulmonary fibrosis 370,371. AM152 is the only LPA1 antagonist currently in clinical trials.
Ki16425 is a LPA1/3 receptor antagonist that demonstrates specificity for LPA1/3, little to no action on LPA2, with no antagonism of S1PR 372. It is widely used in in vitro studies of LPAR function in different disease states, including various cancers, formation of neointima and atherosclerosis development, and fibrosis 161,216,373-375.
VPC12249 is a competitive LPA1/3 antagonist, the development of which resulted from a series of LPAR agonists and antagonists evolving from an agonist parent compound, N-acyl ethanolamide phosphate (NAEPA) and involves the 2-substitution of a bulky benzyl-4-oxybenzyl substituent in the S configuration 376. This compound blocked Ca2+ mobilization and inhibited forskolin-induced cAMP accumulation in HEK293T cells 376. It demonstrates similar effects to Ki6425, inhibiting fibroblast chemotaxis after bleomycin administration and chemotaxis of human pancreatic cancer cells 161,377.
10.2.2 LPAR agonist
OMPT (1-oleoyl-2-O-methyl-rac-glycerophosphothionate) is a LPA 18:1 analogue with the hydroxy group converted to an O-methoxy group, and the phosphate to a phosphothionate, resulting in potent and selective agonism of LPA3 378. Administration of OMPT to mice before induction of renal ischemia/reperfusion injury resulted in exacerbated pathology and was able to block the in vivo protective effects of VPC12249 administration 379.
10.3. S1PR agonists and antagonists
10.3.1 S1PR agonists
FTY720 (Fingolimod/Gilenya; Novartis) is the first LP receptor modulator approved for clinical use. It is a novel oral drug approved by the FDA in September 2010 for the treatment of relapsing-remitting multiple sclerosis 10,380. FTY720 is phosphorylated in vivo by Sphk2 to the active form, FTY720-P (FTY-P) 381-383. Although FTY-P is an agonist of S1P1 and S1P3-5, binding of FTY-P to S1P1 results in down-modulation and degradation of this receptor, amounting to functional antagonism 145,384.
Originally, FTY720 was targeted for use as an anti-rejection treatment in renal transplants; however, clinical trials were halted when it was determined that it provided no increase in prophylaxis of transplant rejection, but did lead to adverse side effects in some patients, including macular edema, bradycardia, reduced renal arterial function, and effects on the pulmonary vasculature 385-387. The apparently immunosuppressive actions of the drug prompted researchers to examine efficacy in experimental autoimmune encephalomyelitis (EAE), a model of human MS, and found that it prevented pathology at concentrations several fold lower than those required for prevention of transplant allografts 382. The precise mechanism of FTY720 in MS is still debated: the sequestration of autoreactive cells in the LN has been considered the primary mechanism of action. However, some studies support the involvement of direct effects on the nervous system itself, including discrepancies between the timing of lymphopenia and amelioration/restoration of symptoms and the accumulation of FTY720 and FTYP in brain tissue 388,389. S1P1 and S1P3 protein up-regulation was found on lesional astrocytes of MS patients, and cell-specific deletion of S1P1 in astrocytes ameliorated EAE pathology to a similar extent as FTY720 administration 313,390. For in-depth discussion of the putative mechanisms of FTY720 in EAE/MS, we refer you to several excellent reviews on the subject that have recently been published: 10,391-393.
FTY720 has shown efficacy in the prevention or amelioration in other models of disease, such as airway inflammation, atherosclerosis, renal ischemia/reperfusion injury, dermal allergic responses, models of Type-1 and -2 diabetes, and autoimmune uveitis 183,185,209,265,394-396. Future studies will likely examine the ability of FTY720 to modulate many more disease pathologies, as well as help determine the potential role of the S1PR in such systems.
AUY954 is a selective S1P1 receptor modulator and an aminocarboxylate analog of FTY720, effective at low nanomolar concentrations 397. Thus, it is a useful tool to dissect the effects of FTY720, or to attribute outcomes specifically to agonism of S1P1 113,182,246,313. However, it also induces receptor internalization and degradation and therefore also acts as a functional antagonist 398. Administration of AUY954 in EAE and models of airway inflammation or autoimmune neuritis recapitulates the effects seen with FTY720 182,313,399.
SEW2871 is a reversible, moderate-affinity, S1P1-specific agonist that binds to the same site as S1P 400,401. Because of this specificity, unlike FTY720, it does not induce deleterious side effects that result from agonism of S1P3, such as bradycardia 133. It also resembles S1P in that it induces signaling and receptor internalization without degradation, also unlike FTY720401. Although long-term administration results in lymphopenia resembling that induced by FTY720 treatment, the potency and duration of action of SEW2871 are not as prolonged as the effects of FTY720 402.
10.3.2 S1PR antagonist
As the only known compound with S1P2 specificity, agonistic or antagonistic, JTE-013 has been a critical tool for the dissection of S1P2 inhibitory effects on migration and the complex G-protein signaling pathways involved 403. It has also allowed for assessing the relative contribution of S1P2 activation in vivo, such as the regulation of B cell homeostasis and in models of atherosclerosis 200,208,288,404. Although JTE-013 has been regarded as a S1P2-specific antagonist, the S1P2 specificity of the compound has recently been questioned 138,405. Ex vivo studies utilizing basilar arteries demonstrated that JTE-013 inhibited vascular contraction induced by not only S1P, but also KCl and a thromboxane analogue, a result obtained using arteries from both wild-type and S1p2-/- mice 131. Additionally, Long, et al. reported that JTE-013 inhibited S1P-induced ERK-1/2 phosphorylation in MDA-MB-453 breast cancer cells, a result reiterated by S1P4, but not S1P2 knockdown, indicating that JTE-013 may be better classified as an S1P2/4 dual antagonist 406.
11. Receptor-independent actions of S1P and LPA
Although the signaling events downstream of S1PR and LPAR ligand binding have been extensively examined, S1P and LPA were originally proposed to transduce non-receptor-mediated signals. Early studies indicated that S1P and precursor molecule ceramide were important second messengers in multiple mitogenesis and apoptosis pathways 407,408,409,410,411. Likewise, LPA was a known inducer of platelet activation and vasopression, via signaling of intracellularly synthesized LPA or modulation of cellular membrane architecture, respectively 412,413. Although later studies would demonstrate the role of GPCR signaling in these effects, others appear to be initiated, in part or whole, directly by intracellular LPs 56,101,412,414,415,416.
One such effect is the release of intracellular Ca2+ stores. Although activation of some LPAR and S1PR can lead to intracellular Ca2+ mobilization, intracellular LPA or S1P are capable of eliciting Ca2+ flux via store-operated or non-store-operated Ca2+ entry channels in diverse cell types, such as neutrophils, mast cells, macrophages, vascular smooth muscle cells, and pulmonary endothelial cells 46,171,172,417-420. Whether such effects are direct signaling function of the LPs on channel proteins or indirectly through membrane effects is not known. Recent in vitro studies have identified intracellular S1P as a negative regulator of histone deacetylase activity, a necessary component of the NF-kB activation cascade via activation of the E3 ubiquitin ligase TRAF2, a modulator of Rhodopsin 1 endosomal trafficking, and is necessary for assembly of the electron transport chain and mitochondrial function 421-424. PPARγ (peroxisome proliferator-activated receptor γ) agonism as a result of binding LPA was demonstrated in RAW264.7 monocytic cells and CHO cells, and in vivo using LPAR knockout mice in a model of chemically-induced arterial wall remodeling 425-427. The relevance of such interactions to physiology and disease is not clear at present.
In some cases, the physiological contributions ascribed to non-GPCR-mediated S1P or LPA signaling are still unclear, due to the difficulties in differentiating between true second messenger signaling versus intracellular synthesis and subsequent autocrine or transcellular GPCR activation. For instance, mast cells produce and secrete S1P, which is thought to bind S1PR to modulate FcεR-mediated degranulation and inflammatory mediator synthesis, and in vivo studies using Sphk1-/- or Sphk2-/- mice yield conflicting results 46,428,429. Similarly, in studies utilizing preadipocytes, LPA binding to LPA1 repressed PPARγ in vitro, and mice with adipocyte-specific deletion of ATX demonstrated increased adipocity with a concomitant increase in PPARγ expression, implying that the LPA that affects PPARγ is produced extracellularly and signals via LPAR430,431. Additionally, there have been reports of intracellular expression of LPA1 and S1P5 432,433. Thus, disentangling the relative contributions of GPCR- versus non-GPCR-mediated regulation of some cellular responses to either LPA or S1P will require meticulous dissection in both in vitro and in vivo systems.
12. Conclusions and perspectives
In the current review, we have attempted to comprehensively address the current state of our understanding of all bona fide members of the S1P and LPA receptor families, with emphasis on their contribution to the regulation of specific cell types in various systems. The past decade has seen a vigorous expansion in the study of S1P and LPA receptors. An increasing repertoire of agonists and antagonists, as well as the development of knockout and knockin animal models, has lead to our appreciation of these receptors as critical players in the regulation of homeostatic and disease states of most of the major body systems. Although the initial foci of S1PR and LPAR studies were the vascular or nervous systems, respectively, oncology, reproduction, immunology, and stem cell biology are just a few of the other fields that these receptors have been shown to dramatically influence.
Despite our increasing knowledge, the complexity and heterogeneity of LP receptor expression and signaling patterns demands intense investigation to fully comprehend the regulatory networks controlled by these receptors. New pathways and networks will likely emerge with further interrogation of the less-well characterized receptors, S1P4 and S1P5, and LPA4-6, expanding the known overlapping or unique functionality of each family and individual receptor. The recent FDA approval of the S1PR modulator FTY720 as a first-line treatment for MS has heightened interest in the field of lysophospholipids and their receptors, and is likely a prelude to increased research and the future introduction of other LP receptor modulators into therapeutic use.
Figure 1. Metabolism of LPA, LPAR G-protein coupling and down-stream signaling pathways.

PC, phosphatidylcholine; PE, phosphoethanolamine; PS, phosphatidylserine; PLD, phospholipase D; PLA1/2, phospholipase A1/2; PA, phosphatidic acid; LPC, lysophosphatidylcholine; LPA, lysophosphatidic acid; MAG, monoacylglycerol; LPP, lipid phosphate phosphatase; LDL, low-density lipoprotein; ATX/lysoPLD, autotaxin/lysophospholipase D; AC, adenyl cyclase; cAMP, cyclic adenosine monophosphate; ROCK, RhoA-associated kinase; SRF, serum response factor; PLC, phospholipase C; PKC, protein kinase C; PI3K, phosphoinositide 3-kinase; MAPK, mitogen-activated protein kinase; eNOS, endothelial nitric oxide synthase. Dotted red line indicates suggested LPAR-G-protein coupling pathway.
Acknowledgments
We gratefully acknowledge the support of the NIH, grants F32CA142117 (V.A.B.) and HL89934 and HL70694 (T.H.).
Biographies

Victoria Blaho obtained her undergraduate B.S. in biochemistry from the University of Missouri. She completed her graduate studies in the laboratory of Dr. Charles Brown at the University of Missouri and received a Ph.D. in Molecular Microbiology and Immunology in 2007. She is currently a post-doctoral scientist mentored by Dr. Timothy Hla at Weill Cornell Medical College. Her current studies include examining the role of the sphingosine 1-phosphate receptors in the regulation of immune cell homeostasis.

Timothy Hla is a Professor of Pathology and Laboratory Medicine and the Director of the Center for Vascular Biology at the Weill Cornell Medical College, Cornell University in New York City. He received his Ph.D. in Biochemistry at the George Washington University in Washington, D.C. and did his postdoc with Thomas Maciag at the American Red Cross Holland Laboratory, Rockville, MD. His major contributions include the molecular characterization of human cyclooxygenase-2 enzyme, the discovery of the sphingosine 1-phosphate receptor and the elucidation of vascular effects of bioactive lipids. His laboratory continues to investigate the vascular biology of bioactive lysophospholipids and the role of RNA binding proteins in gene expression and cellular behavior.
References
- 1.Lynch KR, Macdonald TL. Prostaglandins Other Lipid Mediat. 2001;64:33. doi: 10.1016/s0090-6980(01)00106-x. [DOI] [PubMed] [Google Scholar]
- 2.Hannun YA, Obeid LM. Nat Rev Mol Cell Biol. 2008;9:139. doi: 10.1038/nrm2329. [DOI] [PubMed] [Google Scholar]
- 3.Chun J, Hla T, Lynch KR, Spiegel S, Moolenaar WH. Pharmacol Rev. 2010;62:579. doi: 10.1124/pr.110.003111. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Tigyi G, Parrill AL. Prog Lipid Res. 2003;42:498. doi: 10.1016/s0163-7827(03)00035-3. [DOI] [PubMed] [Google Scholar]
- 5.Lynch KR, Macdonald TL. Biochim Biophys Acta. 2002;1582:289. doi: 10.1016/s1388-1981(02)00183-x. [DOI] [PubMed] [Google Scholar]
- 6.Tokumura A, Sinomiya J, Kishimoto S, Tanaka T, Kogure K, Sugiura T, Satouchi K, Waku K, Fukuzawa K. Biochem J. 2002;365:617. doi: 10.1042/BJ20020348. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Murakami-Murofushi K, Uchiyama A, Fujiwara Y, Kobayashi T, Kobayashi S, Mukai M, Murofushi H, Tigyi G. Biochim Biophys Acta. 2002;1582:1. doi: 10.1016/s1388-1981(02)00131-2. [DOI] [PubMed] [Google Scholar]
- 8.Im D. J Lipid Res. 2004;45:410. doi: 10.1194/jlr.R300006-JLR200. [DOI] [PubMed] [Google Scholar]
- 9.Buzard D, Thatte J, Lerner M, Edwards J. Expert Opin Ther Patents. 2008;18:1141. [Google Scholar]
- 10.Brinkmann V, Billich A, Baumruker T, Heining P, Schmouder R, Francis G, Aradhye S, Burtin P. Nat Rev Drug Discov. 2010;9:883. doi: 10.1038/nrd3248. [DOI] [PubMed] [Google Scholar]
- 11.Hecht JH, Weiner JA, Post SR, Chun J. J Cell Biol. 1996;135:1071. doi: 10.1083/jcb.135.4.1071. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Hla T, Maciag T. J Biol Chem. 1990;265:9308. [PubMed] [Google Scholar]
- 13.Lee MJ, Van Brocklyn JR, Thangada S, Liu CH, Hand AR, Menzeleev R, Spiegel S, Hla T. Science. 1998;279:1552. doi: 10.1126/science.279.5356.1552. [DOI] [PubMed] [Google Scholar]
- 14.Pamuklar Z, Federico L, Liu S, Umezu-Goto M, Dong A, Panchatcharam M, Fulkerson Z, Fulerson Z, Berdyshev E, Natarajan V, Fang X, van Meeteren LA, Moolenaar WH, Mills GB, Morris AJ, Smyth SS. J Biol Chem. 2009;284:7385. doi: 10.1074/jbc.M807820200. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.van Meeteren LA, Moolenaar WH. Prog Lipid Res. 2007;46:145. doi: 10.1016/j.plipres.2007.02.001. [DOI] [PubMed] [Google Scholar]
- 16.Fyrst H, Saba JD. Nat Chem Bio. 2010;6:489. doi: 10.1038/nchembio.392. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Venkataraman K, Thangada S, Michaud J, Oo ML, Ai Y, Lee YM, Wu M, Parikh NS, Khan F, Proia RL, Hla T. Biochem J. 2006;397:461. doi: 10.1042/BJ20060251. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Tanaka M, Okudaira S, Kishi Y, Ohkawa R, Iseki S, Ota M, Noji S, Yatomi Y, Aoki J, Arai H. J Biol Chem. 2006;281:25822. doi: 10.1074/jbc.M605142200. [DOI] [PubMed] [Google Scholar]
- 19.Schwab SR, Pereira JP, Matloubian M, Xu Y, Huang Y, Cyster JG. Science. 2005;309:1735. doi: 10.1126/science.1113640. [DOI] [PubMed] [Google Scholar]
- 20.Baker DL, Desiderio DM, Miller DD, Tolley B, Tigyi GJ. Anal Biochem. 2001;292:287. doi: 10.1006/abio.2001.5063. [DOI] [PubMed] [Google Scholar]
- 21.Pappu R, Schwab SR, Cornelissen I, Pereira JP, Regard JB, Xu Y, Camerer E, Zheng YW, Huang Y, Cyster JG, Coughlin SR. Science. 2007;316:295. doi: 10.1126/science.1139221. [DOI] [PubMed] [Google Scholar]
- 22.Sano T, Baker D, Virag T, Wada A, Yatomi Y, Kobayashi T, Igarashi Y, Tigyi G. J Biol Chem. 2002;277:21197. doi: 10.1074/jbc.M201289200. [DOI] [PubMed] [Google Scholar]
- 23.Alexander JS, Patton WF, Christman BW, Cuiper LL, Haselton FR. Am J Physiol. 1998;274:H115. doi: 10.1152/ajpheart.1998.274.1.H115. [DOI] [PubMed] [Google Scholar]
- 24.Hänel P, Andréani P, Gräler MH. F A S E B J. 2007;21:1202. doi: 10.1096/fj.06-7433com. [DOI] [PubMed] [Google Scholar]
- 25.Moolenaar WH. Curr Opin Cell Biol. 1995;7:203. doi: 10.1016/0955-0674(95)80029-8. [DOI] [PubMed] [Google Scholar]
- 26.Venkataraman K, Lee YM, Michaud J, Thangada S, Ai Y, Bonkovsky HL, Parikh NS, Habrukowich C, Hla T. Circ Res. 2008;102:669. doi: 10.1161/CIRCRESAHA.107.165845. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Pham THM, Baluk P, Xu Y, Grigorova I, Bankovich AJ, Pappu R, Coughlin SR, McDonald DM, Schwab SR, Cyster JG. J Exp Med. 2010;207:17. doi: 10.1084/jem.20091619. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Savaskan NE, Rocha L, Kotter MR, Baer A, Lubec G, van Meeteren LA, Kishi Y, Aoki J, Moolenaar WH, Nitsch R, Bräuer AU. Cell Mol Life Sci. 2007;64:230. doi: 10.1007/s00018-006-6412-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Ferry G, Tellier E, Try A, Grés S, Naime I, Simon MF, Rodriguez M, Boucher J, Tack I, Gesta S, Chomarat P, Dieu M, Raes M, Galizzi JP, Valet P, Boutin JA, Saulnier-Blache JS. J Biol Chem. 2003;278:18162. doi: 10.1074/jbc.M301158200. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Huang MC, Graeler M, Shankar G, Spencer J, Goetzl EJ. Biochem Biophys Acta. 2002;1582:161. doi: 10.1016/s1388-1981(02)00151-8. [DOI] [PubMed] [Google Scholar]
- 31.Fang X, Gaudette D, Furui T, Mao M, Estrella V, Eder A, Pustilnik T, Sasagawa T, Lapushin R, Yu S, Jaffe RB, Wiener JR, Erickson JR, Mills GB. Ann N Y Acad Sci. 2000;905:188. doi: 10.1111/j.1749-6632.2000.tb06550.x. [DOI] [PubMed] [Google Scholar]
- 32.Yang L, Yatomi Y, Miura Y, Satoh K, Ozaki Y. Br J Haematol. 1999;107:282. doi: 10.1046/j.1365-2141.1999.01697.x. [DOI] [PubMed] [Google Scholar]
- 33.Aoki J, Taira A, Takanezawa Y, Kishi Y, Hama K, Kishimoto T, Mizuno K, Saku K, Taguchi R, Arai H. J Biol Chem. 2002;277:48737. doi: 10.1074/jbc.M206812200. [DOI] [PubMed] [Google Scholar]
- 34.Kohno M, Momoi M, Oo ML, Paik JH, Lee YM, Venkataraman K, Ai Y, Ristimaki AP, Fyrst H, Sano H, Rosenberg D, Saba JD, Proia RL, Hla T. Mol Cell Biol. 2006;26:7211. doi: 10.1128/MCB.02341-05. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Cummings R, Parinandi N, Wang L, Usatyuk P, Natarajan V. Mol Cell Biochem. 2002:234–235. 99. [PubMed] [Google Scholar]
- 36.Jeng YJ, Suarez VR, Izban MG, Wang HQ, Soloff MS. Am J Physiol Endocrinol Metab. 2006;292:E1110. doi: 10.1152/ajpendo.00373.2006. [DOI] [PubMed] [Google Scholar]
- 37.Fukuda Y. Biochem Biophys Res Comm. 2003;309:155. doi: 10.1016/s0006-291x(03)01551-1. [DOI] [PubMed] [Google Scholar]
- 38.Ye X. Human Reproduction Update. 2008;14:519–536. doi: 10.1093/humupd/dmn023. [DOI] [PubMed] [Google Scholar]
- 39.Kim M, Park SW, Kim M, D'Agati VD, Lee HT. Anesthesiology. 2011;114:363. doi: 10.1097/ALN.0b013e3182070c3a. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Fukushima N, Weiner JA, Kaushal D, Contos JJA, Rehen SK, Kingsbury MA, Kim KY, Chun J. Mol Cell Neurosci. 2002;20:271. doi: 10.1006/mcne.2002.1123. [DOI] [PubMed] [Google Scholar]
- 41.D'Arrigo P, Servi S. Molecules. 2010;15:1354. doi: 10.3390/molecules15031354. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.van Meeteren LA, Ruurs P, Stortelers C, Bouwman P, van Rooijen MA, Pradère JP, Pettit TR, Wakelam MJO, Saulnier-Blache JS, Mummery CL, Moolenaar WH, Jonkers J. Mol Cell Biol. 2006;26:5015. doi: 10.1128/MCB.02419-05. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Kohama T, Olivera A, Edsall L, Nagiec MM, Dickson R, Spiegel S. J Biol Chem. 1998;273:23722. doi: 10.1074/jbc.273.37.23722. [DOI] [PubMed] [Google Scholar]
- 44.Liu H, Sugiura M, Nava VE, Edsall LC, Kono K, Poulton S, Milstien S, Kohama T, Spiegel S. J Biol Chem. 2000;275:19513. doi: 10.1074/jbc.M002759200. [DOI] [PubMed] [Google Scholar]
- 45.Mizugishi K, Yamashita T, Olivera A, Miller GF, Spiegel S, Proia RL. Mol Cell Biol. 2005;25:11113. doi: 10.1128/MCB.25.24.11113-11121.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Olivera A, Mizugishi K, Tikhonova A, Ciaccia L, Odom S, Proia RL, Rivera J. Immunity. 2007;26:287. doi: 10.1016/j.immuni.2007.02.008. [DOI] [PubMed] [Google Scholar]
- 47.Olivera A, Eisner C, Kitamura Y, Dillahunt S, Allende L, Tuymetova G, Watford W, Meylan F, Diesner SC, Li L, Schnermann J, Proia RL, Rivera J. J Clin Invest. 2010;120:1429. doi: 10.1172/JCI40659. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Allende ML, Sasaki T, Kawai H, Olivera A, Mi Y, van Echten-Deckert G, Hajdu R, Rosenbach M, Keohane CA, Mandala S, Spiegel S, Proia RL. J Biol Chem. 2004;279:52487. doi: 10.1074/jbc.M406512200. [DOI] [PubMed] [Google Scholar]
- 49.Sensken SC, Bode C, Nagarajan M, Peest U, Pabst O, Gräler MH. J Immunol. 2010;184:4133. doi: 10.4049/jimmunol.0903358. [DOI] [PubMed] [Google Scholar]
- 50.Michaud J, Kohno M, Proia RL, Hla T. F E B S Letters. 2006;580:4607. doi: 10.1016/j.febslet.2006.07.035. [DOI] [PubMed] [Google Scholar]
- 51.Smyth SS, Sciorra VA, Sigal YJ, Pamuklar Z, Wang Z, Xu Y, Prestwich GD, Morris AJ. J Biol Chem. 2003;278:43214. doi: 10.1074/jbc.M306709200. [DOI] [PubMed] [Google Scholar]
- 52.Sigal YJ, McDermott MI, Morris AJ. Biochem J. 2005;387:281. doi: 10.1042/BJ20041771. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Yamashita A, Kawagishi N, Miyashita T, Nagatsuka T, Sugiura T, Kume K, Shimizu T, Waku K. J Biol Chem. 2001;276:26745. doi: 10.1074/jbc.M101795200. [DOI] [PubMed] [Google Scholar]
- 54.Zhou J, Saba JD. Biochem Biophys Res Commun. 1998;242:502. doi: 10.1006/bbrc.1997.7993. [DOI] [PubMed] [Google Scholar]
- 55.Ito K, Anada Y, Tani M, Ikeda M, Sano T, Kihara A, Igarashi Y. Biochem Biophys Res Comm. 2007;357:212. doi: 10.1016/j.bbrc.2007.03.123. [DOI] [PubMed] [Google Scholar]
- 56.Yatomi Y, Yamamura S, Ruan F, Igarashi Y. J Biol Chem. 1997;272:5291. doi: 10.1074/jbc.272.8.5291. [DOI] [PubMed] [Google Scholar]
- 57.Mechtcheriakova D, Wlachos A, Sobanov J, Kopp T, Reuschel R, Bornancin F, Cai R, Zemann B, Urtz N, Stingl G, Zlabinger G, Woisetschläger M, Baumruker T, Billich A. Cell Signal. 2007;19:748. doi: 10.1016/j.cellsig.2006.09.004. [DOI] [PubMed] [Google Scholar]
- 58.Pham TCT, Fells JI, Osborne DA, North EJ, Naor MM, Parrill AL. J Mol Graph Model. 2008;26:1189. doi: 10.1016/j.jmgm.2007.11.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Yin H, Chu A, Li W, Wang B, Shelton F, Otero F, Nguyen DG, Caldwell JS, Chen YA. J Biol Chem. 2009;284:12328. doi: 10.1074/jbc.M806516200. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Lee MJ, Thangada S, Liu CH, Thompson BD, Hla T. J Biol Chem. 1998;273:22105. doi: 10.1074/jbc.273.34.22105. [DOI] [PubMed] [Google Scholar]
- 61.Wang Da, Lorincz Z, Bautista DL, Liliom K, Tigyi G, Parrill AL. J Biol Chem. 2001;276:49213. doi: 10.1074/jbc.M107301200. [DOI] [PubMed] [Google Scholar]
- 62.Fujiwara Y, Osborne DA, Walker MD, Wang Da, Bautista DA, Liliom K, Van Brocklyn JR, Parrill AL, Tigyi G. J Biol Chem. 2007;282:2374. doi: 10.1074/jbc.M609648200. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Choi JW, Herr DR, Noguchi K, Yung YC, Lee CW, Mutoh T, Lin ME, Teo ST, Park KE, Mosley AN, Chun J. Ann Rev Pharmicol Toxicol. 2010;50:157. doi: 10.1146/annurev.pharmtox.010909.105753. [DOI] [PubMed] [Google Scholar]
- 64.Hla T. Prostaglandins Other Lipid Med. 2005;77:197. doi: 10.1016/j.prostaglandins.2005.06.008. [DOI] [PubMed] [Google Scholar]
- 65.Mitra P, Oskeritzian CA, Payne SG, Beaven MA, Milstien S, Spiegel S. Proc Nat Acad Sci U S A. 2006;103:16394. doi: 10.1073/pnas.0603734103. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Kobayashi N, Nishi T, Hirata T, Kihara A, Sano T, Igarashi Y, Yamaguchi A. J Lipid Res. 2006;47:614. doi: 10.1194/jlr.M500468-JLR200. [DOI] [PubMed] [Google Scholar]
- 67.Kobayashi N, Kobayashi N, Yamaguchi A, Nishi T. J Biol Chem. 2009;284:21192. doi: 10.1074/jbc.M109.006163. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Lee Y, Venkataraman K, Hwang S, Han D, Hla T. Prostaglandins Other Lipid Med. 2007;84:154. doi: 10.1016/j.prostaglandins.2007.08.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Kamiyama E, Sugiyama D, Nakai D, Miura Si, Okazaki O. Drug Metab Dispos. 2009;37:1956. doi: 10.1124/dmd.109.027490. [DOI] [PubMed] [Google Scholar]
- 70.Kirchheiner J, Brockmöller J, Meineke I, Bauer S, Rohde W, Meisel C, Roots I. Clin Pharmacol Ther. 2002;71:286. doi: 10.1067/mcp.2002.122476. [DOI] [PubMed] [Google Scholar]
- 71.Kawahara A, Nishi T, Hisano Y, Fukui H, Yamaguchi A, Mochizuki N. Science. 2009;323:524. doi: 10.1126/science.1167449. [DOI] [PubMed] [Google Scholar]
- 72.Hisano Y, Kobayashi N, Kawahara A, Yamaguchi A, Nishi T. J Biol Chem. 2011;286:1758. doi: 10.1074/jbc.M110.171116. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Takabe K, Kim RH, Allegood JC, Mitra P, Ramachandran S, Nagahashi M, Harikumar KB, Hait NC, Milstien S, Spiegel S. J Biol Chem. 2010;285:10477. doi: 10.1074/jbc.M109.064162. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Sato K, Malchinkhuu E, Horiuchi Y, Mogi C, Tomura H, Tosaka M, Yoshimoto Y, Kuwabara A, Okajima F. J Neurochem. 2007;103:2610. doi: 10.1111/j.1471-4159.2007.04958.x. [DOI] [PubMed] [Google Scholar]
- 75.Jones TR, Zamboni R, Belley M, Champion E, Charette L, Ford-Hutchinson AW, Frenette R, Gauthier JY, Leger S, Masson P. Can J Physiol Pharmacol. 1989;67:17. doi: 10.1139/y89-004. [DOI] [PubMed] [Google Scholar]
- 76.Brink C, Dahlen SE, Drazen J, Evans JF, Hay DWP, Nicosia S, Serhan CN, Shimizu T, Yokomizo T. Pharmacol Rev. 2003;55:195. doi: 10.1124/pr.55.1.8. [DOI] [PubMed] [Google Scholar]
- 77.Osborne N, Brand-Arzamendi K, Ober EA, Jin SW, Verkade H, Holtzman NG, Yelon D, Stainier DYR. Curr Biol. 2008;18:1882. doi: 10.1016/j.cub.2008.10.061. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Rader DJ, Daugherty A. Nature. 2008;451:904. doi: 10.1038/nature06796. [DOI] [PubMed] [Google Scholar]
- 79.Park YM, Febbraio M, Silverstein RL. J Clin Invest. 2009;119:136. doi: 10.1172/JCI35535. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Miller M, Zhan M. Curr Opin Cardiol. 2004;19:380. doi: 10.1097/01.hco.0000126584.12520.b5. [DOI] [PubMed] [Google Scholar]
- 81.Fueller M, Wang Da, Tigyi G, Siess W. Cell Signal. 2003;15:367. doi: 10.1016/s0898-6568(02)00117-1. [DOI] [PubMed] [Google Scholar]
- 82.Siess W, Zangl KJ, Essler M, Bauer M, Brandl R, Corrinth C, Bittman R, Tigyi G, Aepfelbacher M. Proc Nat Acad Sci U S A. 1999;96:6931. doi: 10.1073/pnas.96.12.6931. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Siess W, Essler M, Brandl R. I U B M B Life. 2000;49:167. doi: 10.1080/713803618. [DOI] [PubMed] [Google Scholar]
- 84.Kimura T, Sato K, Kuwabara A, Tomura H, Ishiwara M, Kobayashi I, Ui M, Okajima F. J Biol Chem. 2001;276:31780. doi: 10.1074/jbc.M104353200. [DOI] [PubMed] [Google Scholar]
- 85.Kimura T, Sato K, Malchinkhuu E, Tomura H, Tamama K, Kuwabara A, Murakami M, Okajima F. Arterioscler Thromb Vasc Biol. 2003;23:1283. doi: 10.1161/01.ATV.0000079011.67194.5A. [DOI] [PubMed] [Google Scholar]
- 86.Murata N, Sato K, Kon J, Tomura H, Yanagita M, Kuwabara A, Ui M, Okajima F. Biochem J. 2000;352:809. [PMC free article] [PubMed] [Google Scholar]
- 87.Argraves KM, Argraves WS. J Lipid Res. 2007;48:2325. doi: 10.1194/jlr.R700011-JLR200. [DOI] [PubMed] [Google Scholar]
- 88.Christoffersen C, Obinata H, Kumaraswamy SB, Galvani S, Ahnström J, Sevvana M, Egerer-Sieber C, Muller YA, Hla T, Nielsen LB, Dahlbäck B. Proc Natl Acad Sci U S A. 2011;108:9613. doi: 10.1073/pnas.1103187108. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Hansson GK, Robertson AKL, Söderberg-Nauclér C. Ann Rev Pathol. 2006;1:297. doi: 10.1146/annurev.pathol.1.110304.100100. [DOI] [PubMed] [Google Scholar]
- 90.Kanda H, Newton R, Klein R, Morita Y, Gunn MD, Rosen SD. Nat Immunol. 2008;9:415. doi: 10.1038/ni1573. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Hausmann J, Kamtekar S, Christodoulou E, Day JE, Wu T, Fulkerson Z, Albers HMHG, van Meeteren LA, Houben AJS, van Zeijl L, Jansen S, Andries M, Hall T, Pegg LE, Benson TE, Kasiem M, Harlos K, Kooi CWV, Smyth SS, Ovaa H, Bollen M, Morris AJ, Moolenaar WH, Perrakis A. Nat Struct Mol Biol. 18:198. doi: 10.1038/nsmb.1980. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Nishimasu H, Okudaira S, Hama K, Mihara E, Dohmae N, Inoue A, Ishitani R, Takagi J, Aoki J, Nureki O. Nat Struct Mol Biol. 2011;18:205. doi: 10.1038/nsmb.1998. [DOI] [PubMed] [Google Scholar]
- 93.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. J Clin Invest. 2000;106:951. doi: 10.1172/JCI10905. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Kono M, Mi Y, Liu Y, Sasaki T, Allende ML, Wu YP, Yamashita T, Proia RL. J Biol Chem. 2004;279:29367. doi: 10.1074/jbc.M403937200. [DOI] [PubMed] [Google Scholar]
- 95.Allende ML, Yamashita T, Proia RL. Blood. 2003;102:3665. doi: 10.1182/blood-2003-02-0460. [DOI] [PubMed] [Google Scholar]
- 96.Lee MJ, Thangada S, Claffey KP, Ancellin N, Liu CH, Kluk M, Volpi M, Sha'afi RI, Hla T. Cell. 1999;99:301. doi: 10.1016/s0092-8674(00)81661-x. [DOI] [PubMed] [Google Scholar]
- 97.Thangada S, Paik J, Sapkota G, Ancellin N. Molec Cell. 2001;8:693. doi: 10.1016/s1097-2765(01)00324-0. [DOI] [PubMed] [Google Scholar]
- 98.Takashima SI, Sugimoto N, Takuwa N, Okamoto Y, Yoshioka K, Takamura M, Takata S, Kaneko S, Takuwa Y. Cardiovasc Res. 2008;79:689. doi: 10.1093/cvr/cvn118. [DOI] [PubMed] [Google Scholar]
- 99.Windh RT, Lee MJ, Hla T, An S, Barr AJ, Manning DR. J Biol Chem. 1999;274:27351. doi: 10.1074/jbc.274.39.27351. [DOI] [PubMed] [Google Scholar]
- 100.Lee MJ, Evans M, Hla T. J Biol Chem. 1996;271:11272. doi: 10.1074/jbc.271.19.11272. [DOI] [PubMed] [Google Scholar]
- 101.Van Brocklyn JR, Lee MJ, Menzeleev R, Olivera A, Edsall L, Cuvillier O, Thomas DM, Coopman PJ, Thangada S, Liu CH, Hla T, Spiegel S. J Cell Biol. 1998;142:229. doi: 10.1083/jcb.142.1.229. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Means CK, Miyamoto S, Chun J, Brown JH. J Biol Chem. 2008;283:11954. doi: 10.1074/jbc.M707422200. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Camerer E, Regard JB, Cornelissen I, Srinivasan Y, Duong DN, Palmer D, Pham TH, Wong JS, Pappu R, Coughlin SR. J Clin Invest. 2009;119:1871. doi: 10.1172/JCI38575. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Morales-Ruiz M, Lee MJ, Zöllner S, Gratton JP, Scotland R, Shiojima I, Walsh K, Hla T, Sessa WC. J Biol Chem. 2001;276:19672. doi: 10.1074/jbc.M009993200. [DOI] [PubMed] [Google Scholar]
- 105.Krump-Konvalinkova V, Yasuda S, Rubic T, Makarova N, Mages J, Erl W, Vosseler C, Kirkpatrick CJ, Tigyi G, Siess W. Arterioscler Thromb Vasc Biol. 2005;25:546. doi: 10.1161/01.ATV.0000154360.36106.d9. [DOI] [PubMed] [Google Scholar]
- 106.Birker-Robaczewska M, Studer R, Haenig B, Menyhart K, Hofmann S, Nayler O. J Cell Biochem. 2008;105:1139. doi: 10.1002/jcb.21918. [DOI] [PubMed] [Google Scholar]
- 107.Ishii I, Friedman B, Ye X, Kawamura S, McGiffert C, Contos JJA, Kingsbury MA, Zhang G, Brown JH, Chun J. J Biol Chem. 2001;276:33697. doi: 10.1074/jbc.M104441200. [DOI] [PubMed] [Google Scholar]
- 108.Ishii I, Ye X, Friedman B, Kawamura S, Contos JJA, Kingsbury MA, Yang AH, Zhang G, Brown JH, Chun J. J Biol Chem. 2002;277:25152. doi: 10.1074/jbc.M200137200. [DOI] [PubMed] [Google Scholar]
- 109.Liu Z, Tan JL, Cohen DM, Yang MT, Sniadecki NJ, Ruiz SA, Nelson CM, Chen CS. Proc Natl Acad Sci U S A. 2010;107:9944. doi: 10.1073/pnas.0914547107. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.Terry SJ, Zihni C, Elbediwy A, Vitiello E, San IVLC, Balda MS, Matter K. Nat Cell Biol. 2011;13:159. doi: 10.1038/ncb2156. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111.Hoffman BD, Grashoff C, Schwartz MA. Nature. 475:316. doi: 10.1038/nature10316. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112.Singleton PA, Moreno-Vinasco L, Sammani S, Wanderling SL, Moss J, Garcia JGN. Am J Respir Cell Mol Biol. 2007;37:222. doi: 10.1165/rcmb.2006-0327OC. [DOI] [PubMed] [Google Scholar]
- 113.Niessen F, Schaffner F, Furlan-Freguia C, Pawlinski R, Bhattacharjee G, Chun J, Derian CK, Andrade-Gordon P, Rosen H, Ruf W. Nature. 2008;452:654. doi: 10.1038/nature06663. [DOI] [PubMed] [Google Scholar]
- 114.Stockton RA, Shenkar R, Awad IA, Ginsberg MH. J Exp Med. 2010;207:881. doi: 10.1084/jem.20091258. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115.Mong PY, Wang Q. J Immunol. 2009;182:2385. doi: 10.4049/jimmunol.0802811. [DOI] [PubMed] [Google Scholar]
- 116.Singleton PA, Dudek SM, Ma SF, Garcia JGN. J Biol Chem. 2006;281:34381. doi: 10.1074/jbc.M603680200. [DOI] [PubMed] [Google Scholar]
- 117.Sammani S, Moreno-Vinasco L, Mirzapoiazova T, Singleton PA, Chiang ET, Evenoski CL, Wang T, Mathew B, Husain A, Moitra J, Sun X, Nunez L, Jacobson JR, Dudek SM, Natarajan V, Garcia JGN. Am J Respir Cell Mol Biol. 2010;43:394. doi: 10.1165/rcmb.2009-0223OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118.Shea BS, Brooks SF, Fontaine BA, Chun J, Luster AD, Tager AM. Am J Respir Cell Mol Biol. 2010;43:662. doi: 10.1165/rcmb.2009-0345OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119.Sugimoto N, Takuwa N, Okamoto H, Sakurada S, Takuwa Y. Mol Cell Biol. 2003;23:1534. doi: 10.1128/MCB.23.5.1534-1545.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120.Sanchez T, Skoura A, Wu MT, Casserly B, Harrington EO, Hla T. Arterioscler Thromb Vasc Biol. 2007;27:1312. doi: 10.1161/ATVBAHA.107.143735. [DOI] [PubMed] [Google Scholar]
- 121.Kono M, Belyantseva IA, Skoura A, Frolenkov GI, Starost MF, Dreier JL, Lidington D, Bolz SS, Friedman TB, Hla T, Proia RL. J Biol Chem. 2007;282:10690. doi: 10.1074/jbc.M700370200. [DOI] [PubMed] [Google Scholar]
- 122.Lorenz JN, Arend LJ, Robitz R, Paul RJ, MacLennan AJ. Am J Physiol Regul Integr Comp Physiol. 2007;292:R440. doi: 10.1152/ajpregu.00085.2006. [DOI] [PubMed] [Google Scholar]
- 123.Lee JF, Gordon S, Estrada R, Wang L, Siow DL, Wattenberg BW, Lominadze D, Lee MJ. Am J Physiol Heart Circ Physiol. 2008;296:H33. doi: 10.1152/ajpheart.00097.2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124.Zhang G, Xu S, Qian Y, He P. Am J Physiol Heart Circ Physiol. 2010;299:H1494–504. doi: 10.1152/ajpheart.00462.2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125.Ishii S, Nagase T, Shimizu T. Prostaglandins Other Lipid Mediat. 2002;68-69:599. doi: 10.1016/s0090-6980(02)00058-8. [DOI] [PubMed] [Google Scholar]
- 126.Leurs R, Vischer HF, Wijtmans M, de Esch IJP. Trends Pharmacol Sci. 32:250. doi: 10.1016/j.tips.2011.02.004. [DOI] [PubMed] [Google Scholar]
- 127.Curry FRE, Adamson RH. Cardiovasc Res. 2010;87:218. doi: 10.1093/cvr/cvq115. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128.Shimizu T, Nakazawa T, Cho A, Dastvan F, Shilling D, Daum G, Reidy MA. Circ Res. 2007;101:995. doi: 10.1161/CIRCRESAHA.107.159228. [DOI] [PubMed] [Google Scholar]
- 129.Sirois MG, Edelman ER. Am J Physiol. 1997;272:H2746. doi: 10.1152/ajpheart.1997.272.6.H2746. [DOI] [PubMed] [Google Scholar]
- 130.Di Gennaro A, Carnini C, Buccellati C, Ballerio R, Zarini S, Fumagalli F, Viappiani S, Librizzi L, Hernandez A, Murphy RC, Constantin G, De Curtis M, Folco G, Sala A. F A S E B J. 2004;18:842. doi: 10.1096/fj.03-0599fje. [DOI] [PubMed] [Google Scholar]
- 131.Salomone S, Potts EM, Tyndall S, Ip PC, Chun J, Brinkmann V, Waeber C. Br J Pharmacol. 2008;153:140. doi: 10.1038/sj.bjp.0707581. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132.Nofer JR, van der Giet M, Tölle M, Wolinska I, von Wnuck Lipinski K, Baba HA, Tietge UJ, Gödecke A, Ishii I, Kleuser B, Schäfers M, Fobker M, Zidek W, Assmann G, Chun J, Levkau B. J Clin Invest. 2004;113:569. doi: 10.1172/JCI18004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133.Murakami A, Takasugi H, Ohnuma S, Koide Y, Sakurai A, Takeda S, Hasegawa T, Sasamori J, Konno T, Hayashi K, Watanabe Y, Mori K, Sato Y, Takahashi A, Mochizuki N, Takakura N. Mol Pharmacol. 2010;77:704. doi: 10.1124/mol.109.061481. [DOI] [PubMed] [Google Scholar]
- 134.Hemmings DG, Xu Y, Davidge ST. Br J Pharmacol. 2004;143:276. doi: 10.1038/sj.bjp.0705752. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135.Bischoff A, Czyborra P, Meyer Zu Heringdorf D, Jakobs KH, Michel MC. Br J Pharmacol. 2000;130:1878. doi: 10.1038/sj.bjp.0703516. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 136.Salomone S, Yoshimura Si, Reuter U, Foley M, Thomas SS, Moskowitz MA, Waeber C. Eur J Pharmacol. 2003;469:125. doi: 10.1016/s0014-2999(03)01731-x. [DOI] [PubMed] [Google Scholar]
- 137.Szczepaniak WS, Pitt BR, McVerry BJ. Am J Physiol Lung Cell Mol Physiol. 2010;299:L137. doi: 10.1152/ajplung.00233.2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138.Pyne NJ, Pyne S. Front Pharmacol. 2011;2:26. doi: 10.3389/fphar.2011.00026. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 139.Yoon CM, Hong BS, Moon HG, Lim S, Suh PG, Kim YK, Chae CB, Gho YS. Blood. 2008;112:1129. doi: 10.1182/blood-2007-11-125203. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 140.Lee JF, Zeng Q, Ozaki H, Wang L, Hand AR, Hla T, Wang E, Lee MJ. J Biol Chem. 2006;281:29190. doi: 10.1074/jbc.M604310200. [DOI] [PubMed] [Google Scholar]
- 141.Langlois S, Gingras D, Béliveau R. Blood. 2004;103:3020. doi: 10.1182/blood-2003-08-2968. [DOI] [PubMed] [Google Scholar]
- 142.Yonesu K, Kawase Y, Inoue T, Takagi N, Tsuchida J, Takuwa Y, Kumakura S, Nara F. Biochem Pharmacol. 2009;77:1011. doi: 10.1016/j.bcp.2008.12.007. [DOI] [PubMed] [Google Scholar]
- 143.Paik JH, Skoura A, Chae SS, Cowan AE, Han DK, Proia RL, Hla T. Genes Dev. 2004;18:2392. doi: 10.1101/gad.1227804. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 144.Schmid G, Guba M, Ischenko I, Papyan A, Joka M, Schrepfer S, Bruns CJ, Jauch KW, Heeschen C, Graeb C. J Cell Biochem. 2007;101:259. doi: 10.1002/jcb.21181. [DOI] [PubMed] [Google Scholar]
- 145.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. Cancer Res. 2006;66:221. doi: 10.1158/0008-5472.CAN-05-2001. [DOI] [PubMed] [Google Scholar]
- 146.Kupperman E, An S, Osborne N, Waldron S, Stainier DY. Nature. 2000;406:192. doi: 10.1038/35018092. [DOI] [PubMed] [Google Scholar]
- 147.van Nieuw Amerongen GP, Vermeer MA, van Hinsbergh VW. Arterioscler Thromb Vasc Biol. 2000;20:E127. doi: 10.1161/01.atv.20.12.e127. [DOI] [PubMed] [Google Scholar]
- 148.Rivera-Lopez CM, Tucker AL, Lynch KR. Angiogenesis. 2008;11:301. doi: 10.1007/s10456-008-9113-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 149.Lin CI, Chen CN, Lin PW, Chang KJ, Hsieh FJ, Lee H. Biochem Biophys Res Commun. 2007;363:1001. doi: 10.1016/j.bbrc.2007.09.081. [DOI] [PubMed] [Google Scholar]
- 150.Contos JJ, Fukushima N, Weiner JA, Kaushal D, Chun J. Proc Nat Acad Sci U S A. 2000;97:13384. doi: 10.1073/pnas.97.24.13384. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 151.Contos JJA, Ishii I, Fukushima N, Kingsbury MA, Ye X, Kawamura S, Brown JH, Chun J. Mol Cell Biol. 2002;22:6921. doi: 10.1128/MCB.22.19.6921-6929.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 152.Ye X, Hama K, Contos JJA, Anliker B, Inoue A, Skinner MK, Suzuki H, Amano T, Kennedy G, Arai H, Aoki J, Chun J. Nature. 2005;435:104. doi: 10.1038/nature03505. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 153.Lee SJ, Chan TH, Chen TC, Liao BK, Hwang PP, Lee H. The F A S E B J. 2008;22:3706. doi: 10.1096/fj.08-106088. [DOI] [PubMed] [Google Scholar]
- 154.Sumida H, Noguchi K, Kihara Y, Abe M, Yanagida K, Hamano F, Sato S, Tamaki K, Morishita Y, Kano MR, Iwata C, Miyazono K, Sakimura K, Shimizu T, Ishii S. Blood. 2010;116:5060. doi: 10.1182/blood-2010-03-272443. [DOI] [PubMed] [Google Scholar]
- 155.Kim J, Keys JR, Eckhart AD. Cell Signal. 2006;18:1695. doi: 10.1016/j.cellsig.2006.01.009. [DOI] [PubMed] [Google Scholar]
- 156.Lee CW, Rivera R, Dubin AE, Chun J. J Biol Chem. 2007;282:4310. doi: 10.1074/jbc.M610826200. [DOI] [PubMed] [Google Scholar]
- 157.Lee Z, Cheng CT, Zhang H, Subler MA, Wu J, Mukherjee A, Windle JJ, Chen CK, Fang X. Mol Biol Cell. 2008;19:5435. doi: 10.1091/mbc.E08-03-0316. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 158.Ren B, Hale J, Srikanthan S, Silverstein RL. Blood. 2011;117:6036. doi: 10.1182/blood-2010-12-326017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 159.Panchatcharam M, Miriyala S, Yang F, Rojas M, End C, Vallant C, Dong A, Lynch K, Chun J, Morris AJ, Smyth SS. Circ Res. 2008;103:662. doi: 10.1161/CIRCRESAHA.108.180778. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 160.Zhou Z, Niu J, Zhang Z. Mol Biol Rep. 2010;37:2675. doi: 10.1007/s11033-009-9798-6. [DOI] [PubMed] [Google Scholar]
- 161.Tager AM, LaCamera P, Shea BS, Campanella GS, Selman M, Zhao Z, Polosukhin V, Wain J, Karimi-Shah BA, Kim ND, Hart WK, Pardo A, Blackwell TS, Xu Y, Chun J, Luster AD. Nat Med. 2008;14:45. doi: 10.1038/nm1685. [DOI] [PubMed] [Google Scholar]
- 162.Kotarsky K, Boketoft A, Bristulf J, Nilsson NE, Norberg A, Hansson S, Owman C, Sillard R, Leeb-Lundberg LMF, Olde B. J Pharmacol Exp Ther. 2006;318:619. doi: 10.1124/jpet.105.098848. [DOI] [PubMed] [Google Scholar]
- 163.Lee CW, Rivera R, Gardell S, Dubin AE, Chun J. J Biol Chem. 2006;281:23589. doi: 10.1074/jbc.M603670200. [DOI] [PubMed] [Google Scholar]
- 164.Yanagida K, Masago K, Nakanishi H, Kihara Y, Hamano F, Tajima Y, Taguchi R, Shimizu T, Ishii S. J Biol Chem. 2009;284:17731. doi: 10.1074/jbc.M808506200. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 165.Williams JR, Khandoga AL, Goyal P, Fells JI, Perygin DH, Siess W, Parrill AL, Tigyi G, Fujiwara Y. J Biol Chem. 2009;284:17304. doi: 10.1074/jbc.M109.003194. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 166.Boyce BF. Dev Cell. 2009;16:323. doi: 10.1016/j.devcel.2009.03.002. [DOI] [PubMed] [Google Scholar]
- 167.Schwab SR, Cyster JG. Nat Immunol. 2007;8:1295. doi: 10.1038/ni1545. [DOI] [PubMed] [Google Scholar]
- 168.Rosen H, Gonzalez-Cabrera P, Marsolais D, Cahalan S, Don AS, Sanna MG. Immunol Rev. 2008;223:221. doi: 10.1111/j.1600-065X.2008.00645.x. [DOI] [PubMed] [Google Scholar]
- 169.Rivera J, Proia RL, Olivera A. Nat Rev Immunol. 2008;8:753. doi: 10.1038/nri2400. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 170.Nathan C. Nat Rev Immunol. 2006;6:173. doi: 10.1038/nri1785. [DOI] [PubMed] [Google Scholar]
- 171.Itagaki K, Hauser CJ. J Biol Chem. 2003;278:27540. doi: 10.1074/jbc.M301763200. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 172.Itagaki K, Kannan KB, Hauser CJ. J Leukoc Biol. 2005;77:181. doi: 10.1189/jlb.0704390. [DOI] [PubMed] [Google Scholar]
- 173.Rahaman M, Costello RW, Belmonte KE, Gendy SS, Walsh MT. Am J Respir Cell Mol Biol. 2006;34:233. doi: 10.1165/rcmb.2005-0126OC. [DOI] [PubMed] [Google Scholar]
- 174.Aas V, Algerøy S, Sand KL, Iversen JG. Cell Commun Adhes. 2001;8:125. doi: 10.3109/15419060109080712. [DOI] [PubMed] [Google Scholar]
- 175.Sitrin RG, Sassanella TM, Petty HR. Am J Respir Cell Mol Biol. 2011;44:205. doi: 10.1165/rcmb.2010-0019OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 176.Allende ML, Bektas M, Lee BG, Bonifacino E, Kang J, Tuymetova G, Chen W, Saba JD, Proia RL. J Biol Chem. 2011;286:7348. doi: 10.1074/jbc.M110.171819. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 177.Rothenberg ME, Hogan SP. Ann Rev Immunol. 2006;24:147. doi: 10.1146/annurev.immunol.24.021605.090720. [DOI] [PubMed] [Google Scholar]
- 178.Abraham SN, John ALS. Nat Rev Immunol. 2010;10:440. doi: 10.1038/nri2782. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 179.Ammit AJ, Hastie AT, Edsall LC, Hoffman RK, Amrani Y, Krymskaya VP, Kane SA, Peters SP, Penn RB, Spiegel S, Panettieri RA. F A S E B J. 2001;15:1212. doi: 10.1096/fj.00-0742fje. [DOI] [PubMed] [Google Scholar]
- 180.Georas SN. Immunol Res. 2009;45:229. doi: 10.1007/s12026-009-8104-y. [DOI] [PubMed] [Google Scholar]
- 181.Zhao Y, Natarajan V. Cellular signalling. 2009;21:367–377. doi: 10.1016/j.cellsig.2008.10.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 182.Marsolais D, Yagi S, Kago T, Leaf N, Rosen H. Mol Pharmacol. 2011;79:61. doi: 10.1124/mol.110.066811. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 183.Sawicka E, Zuany-Amorim C, Manlius C, Trifilieff A, Brinkmann V, Kemeny DM, Walker C. J Immunol. 2003;171:6206. doi: 10.4049/jimmunol.171.11.6206. [DOI] [PubMed] [Google Scholar]
- 184.Chiba Y, Suzuki K, Kurihara E, Uechi M, Sakai H, Misawa M. Open Respir Med. 2010;4:82. doi: 10.2174/1874306401004010082. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 185.Sugita K, Kabashima K, Sakabe JI, Yoshiki R, Tanizaki H, Tokura Y. Am J Pathol. 2010;177:1881. doi: 10.2353/ajpath.2010.100119. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 186.Mackle T, Gendy SS, Walsh M, McConn-Walsh R, Costello RW, Walsh MT. J Laryngol Otol. 2008;122:1309. doi: 10.1017/S0022215108003241. [DOI] [PubMed] [Google Scholar]
- 187.Roviezzo F, Del Galdo F, Abbate G, Bucci M, D'Agostino B, Antunes E, De Dominicis G, Parente L, Rossi F, Cirino G, De Palma R. Proc Nat Acad Sci U S A. 2004;101:11170. doi: 10.1073/pnas.0401439101. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 188.Idzko M, Laut M, Panther E, Sorichter S, Dürk T, Fluhr JW, Herouy Y, Mockenhaupt M, Myrtek D, Elsner P, Norgauer J. J Immunol. 2004;172:4480. doi: 10.4049/jimmunol.172.7.4480. [DOI] [PubMed] [Google Scholar]
- 189.Muessel MJ, Scott KS, Friedl P, Bradding P, Wardlaw AJ. J Immunol. 2008;180:8354. doi: 10.4049/jimmunol.180.12.8354. [DOI] [PubMed] [Google Scholar]
- 190.Kennerly DA. J Biol Chem. 1987;262:16305. [PubMed] [Google Scholar]
- 191.Choi OH, Kim JH, Kinet JP. Nature. 1996;380:634. doi: 10.1038/380634a0. [DOI] [PubMed] [Google Scholar]
- 192.Bagga S, Price KS, Lin DA, Friend DS, Austen KF, Boyce JA. Blood. 2004;104:4080. doi: 10.1182/blood-2004-03-1166. [DOI] [PubMed] [Google Scholar]
- 193.Lundequist A, Boyce JA. PloS One. 2011;6:e18192. doi: 10.1371/journal.pone.0018192. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 194.Lin DA, Boyce JA. J Immunol. 2005;175:5430. doi: 10.4049/jimmunol.175.8.5430. [DOI] [PubMed] [Google Scholar]
- 195.Hashimoto T, Ohata H, Momose K, Honda K. Pharmacology. 2005;75:13. doi: 10.1159/000085784. [DOI] [PubMed] [Google Scholar]
- 196.Melendez AJ, Khaw AK. J Biol Chem. 2002;277:17255. doi: 10.1074/jbc.M110944200. [DOI] [PubMed] [Google Scholar]
- 197.Jolly PS, Bektas M, Olivera A, Gonzalez-Espinosa C, Proia RL, Rivera J, Milstien S, Spiegel S. J Ex p Med. 2004;199:959. doi: 10.1084/jem.20030680. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 198.Oskeritzian CA, Price MM, Hait NC, Kapitonov D, Falanga YT, Morales JK, Ryan JJ, Milstien S, Spiegel S. J Exp Med. 2010;207:465. doi: 10.1084/jem.20091513. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 199.Michaud J, Im DS, Hla T. J Immunol. 2010;184:1475. doi: 10.4049/jimmunol.0901586. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 200.Skoura A, Michaud J, Im DS, Thangada S, Xiong Y, Smith J, Hla T. Arterioscler Thromb Vasc Biol. 2010;31:81. doi: 10.1161/ATVBAHA.110.213496. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 201.Hughes JE, Srinivasan S, Lynch KR, Proia RL, Ferdek P, Hedrick CC. Circ Res. 2008;102:950. doi: 10.1161/CIRCRESAHA.107.170779. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 202.Hornuβ C, Hammermann R, Fuhrmann M, Juergens UR, Racké K. Eur J Pharmacol. 2001;429:303. doi: 10.1016/s0014-2999(01)01329-2. [DOI] [PubMed] [Google Scholar]
- 203.McQuiston T, Luberto C, Del Poeta M. Microbiology (Reading, England) 2011;157:1416. doi: 10.1099/mic.0.045989-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 204.Lee H, Liao JJ, Graeler M, Huang MC, Goetzl EJ. Biochem Biophys Acta. 2002;1582:175. doi: 10.1016/s1388-1981(02)00153-1. [DOI] [PubMed] [Google Scholar]
- 205.Duong CQ, Bared SM, Abu-Khader A, Buechler C, Schmitz A, Schmitz G. Biochem Biophys Acta. 2004;1682:112. doi: 10.1016/j.bbalip.2004.03.002. [DOI] [PubMed] [Google Scholar]
- 206.Fleit SA, Fleit HB, Zolla-Pazner S. J Immunol Methods. 1984;68:119. doi: 10.1016/0022-1759(84)90142-x. [DOI] [PubMed] [Google Scholar]
- 207.Davies JQ, Gordon S. Methods Mol Biol. 2005;290:91. doi: 10.1385/1-59259-838-2:091. [DOI] [PubMed] [Google Scholar]
- 208.Wang F, Okamoto Y, Inoki I, Yoshioka K, Du W, Qi X, Takuwa N, Gonda K, Yamamoto Y, Ohkawa R, Nishiuchi T, Sugimoto N, Yatomi Y, Mitsumori K, Asano M, Kinoshita M, Takuwa Y. J Clin Invest. 2010;120:3979. doi: 10.1172/JCI42315. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
- 209.Keul P, Lucke S, von Wnuck Lipinski K, Bode C, Gräler M, Heusch G, Levkau B. Circ Res. 2010;108:314. doi: 10.1161/CIRCRESAHA.110.235028. [DOI] [PubMed] [Google Scholar]
- 210.Whetzel AM, Bolick DT, Srinivasan S, Macdonald TL, Morris MA, Ley K, Hedrick CC. Circ Res. 2006;99:731. doi: 10.1161/01.RES.0000244088.33375.52. [DOI] [PubMed] [Google Scholar]
- 211.Barra V, Kuhn AM, von Knethen A, Weigert A, Brüne B. Cell Mol Life Sci. 2011;68:1815. doi: 10.1007/s00018-010-0537-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 212.Weigert A, Johann AM, von Knethen A, Schmidt H, Geisslinger G, Brüne B. Blood. 2006;108:1635. doi: 10.1182/blood-2006-04-014852. [DOI] [PubMed] [Google Scholar]
- 213.Thorp E, Tabas I. J Leukoc Biol. 2009;86:1089. doi: 10.1189/jlb.0209115. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 214.Chang CL, Lin ME, Hsu HY, Yao CL, Hwang SM, Pan CY, Hsu CY, Lee H. J Biomed Sci. 2008;15:357. doi: 10.1007/s11373-007-9223-x. [DOI] [PubMed] [Google Scholar]
- 215.Gustin C, Van Steenbrugge M, Raes M. Am J Physiol Cell Physiol. 2008;295:C905. doi: 10.1152/ajpcell.00544.2007. [DOI] [PubMed] [Google Scholar]
- 216.Zhou Z, Subramanian P, Sevilmis G, Globke B, Soehnlein O, Karshovska E, Megens R, Heyll K, Chun J, Saulnier-Blache JS, Reinholz M, van Zandvoort M, Weber C, Schober A. Cell Metab. 2011;13:592. doi: 10.1016/j.cmet.2011.02.016. [DOI] [PubMed] [Google Scholar]
- 217.Lin CI, Chen CN, Chen JH, Lee H. J Cell Biochem. 2006;99:1216. doi: 10.1002/jcb.20963. [DOI] [PubMed] [Google Scholar]
- 218.Panther E, Idzko M, Corinti S, Ferrari D, Herouy Y, Mockenhaupt M, Dichmann S, Gebicke-Haerter P, Di Virgilio F, Girolomoni G, Norgauer J. J Immunol. 2002;169:4129. doi: 10.4049/jimmunol.169.8.4129. [DOI] [PubMed] [Google Scholar]
- 219.Chen R, Roman J, Guo J, West E, McDyer J, Williams MA, Georas SN. Stem Cells Dev. 2006;15:797. doi: 10.1089/scd.2006.15.797. [DOI] [PubMed] [Google Scholar]
- 220.Chan LC, Peters W, Xu Y, Chun J, Farese RV, Cases S. J Leukoc Biol. 2007;82:1193. doi: 10.1189/jlb.0407221. [DOI] [PubMed] [Google Scholar]
- 221.Gräler MH, Bernhardt G, Lipp M. Genomics. 1998;53:164. doi: 10.1006/geno.1998.5491. [DOI] [PubMed] [Google Scholar]
- 222.Idzko M, Panther E, Corinti S, Morelli A, Ferrari D, Herouy Y, Dichmann S, Mockenhaupt M, Gebicke-Haerter P, Di Virgilio F, Girolomoni G, Norgauer J. F A S E B J. 2002;16:625. doi: 10.1096/fj.01-0625fje. [DOI] [PubMed] [Google Scholar]
- 223.Maeda Y, Matsuyuki H, Shimano K, Kataoka H, Sugahara K, Chiba K. J Immunol. 2007;178:3437. doi: 10.4049/jimmunol.178.6.3437. [DOI] [PubMed] [Google Scholar]
- 224.Gollmann G, Neuwirt H, Tripp CH, Mueller H, Konwalinka G, Heufler C, Romani N, Tiefenthaler M. Internat Immunol. 2008;20:911. doi: 10.1093/intimm/dxn050. [DOI] [PubMed] [Google Scholar]
- 225.Czeloth N, Bernhardt G, Hofmann F, Genth H, Förster R. J Immunol. 2005;175:2960. doi: 10.4049/jimmunol.175.5.2960. [DOI] [PubMed] [Google Scholar]
- 226.Rathinasamy A, Czeloth N, Pabst O, Förster R, Bernhardt G. J Immunol. 2010;185:4072. doi: 10.4049/jimmunol.1000568. [DOI] [PubMed] [Google Scholar]
- 227.Auffray C, Sieweke MH, Geissmann F. Ann Rev Immunol. 2009;27:669. doi: 10.1146/annurev.immunol.021908.132557. [DOI] [PubMed] [Google Scholar]
- 228.Lamana A, Martin P, de la Fuente H, Martinez-Muñoz L, Cruz-Adalia A, Ramirez-Huesca M, Escribano C, Gollmer K, Mellado M, Stein JV, Rodriguez-Fernandez JL, Sanchez-Madrid F, Del Hoyo GM. J Invest Derm. 2011;131:1503. doi: 10.1038/jid.2011.54. [DOI] [PubMed] [Google Scholar]
- 229.Bankovich AJ, Shiow LR, Cyster JG. J Biol Chem. 2010;285:22328. doi: 10.1074/jbc.M110.123299. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 230.Yokoyama WM, Kim S, French AR. Ann Rev Immunol. 2004;22:405. doi: 10.1146/annurev.immunol.22.012703.104711. [DOI] [PubMed] [Google Scholar]
- 231.Jin Y, Knudsen E, Wang L, Maghazachi AA. Eur J Immunol. 2003;33:2083. doi: 10.1002/eji.200323711. [DOI] [PubMed] [Google Scholar]
- 232.Kveberg L, Bryceson Y, Inngjerdingen M, Rolstad B, Maghazachi AA. Eur J Immunol. 2002;32:1856. doi: 10.1002/1521-4141(200207)32:7<1856::AID-IMMU1856>3.0.CO;2-B. [DOI] [PubMed] [Google Scholar]
- 233.Walzer T, Chiossone L, Chaix J, Calver A, Carozzo C, Garrigue-Antar L, Jacques Y, Baratin M, Tomasello E, Vivier E. Nat Immunol. 2007;8:1337. doi: 10.1038/ni1523. [DOI] [PubMed] [Google Scholar]
- 234.Lagadari M, Truta-Feles K, Lehmann K, Berod L, Ziemer M, Idzko M, Barz D, Kamradt T, Maghazachi AA, Norgauer J. Internat Immunol. 2009;21:667. doi: 10.1093/intimm/dxp035. [DOI] [PubMed] [Google Scholar]
- 235.Maghazachi AA. Pharmacol Rev. 2005;57:339. doi: 10.1124/pr.57.3.5. [DOI] [PubMed] [Google Scholar]
- 236.Szabo SJ, Sullivan BM, Stemmann C, Satoskar AR, Sleckman BP, Glimcher LH. Science. 2002;295:338. doi: 10.1126/science.1065543. [DOI] [PubMed] [Google Scholar]
- 237.Jenne CN, Enders A, Rivera R, Watson SR, Bankovich AJ, Pereira JP, Xu Y, Roots CM, Beilke JN, Banerjee A, Reiner SL, Miller SA, Weinmann AS, Goodnow CC, Lanier LL, Cyster JG, Chun J. J Exp Med. 2009;206:2469. doi: 10.1084/jem.20090525. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 238.Kinjo Y, Tupin E, Wu D, Fujio M, Garcia-Navarro R, Benhnia MR, Zajonc DM, Ben-Menachem G, Ainge GD, Painter GF, Khurana A, Hoebe K, Behar SM, Beutler B, Wilson IA, Tsuji M, Sellati TJ, Wong CH, Kronenberg M. Nat Immunol. 2006;7:978. doi: 10.1038/ni1380. [DOI] [PubMed] [Google Scholar]
- 239.Fox LM, Cox DG, Lockridge JL, Wang X, Chen X, Scharf L, Trott DL, Ndonye RM, Veerapen N, Besra GS, Howell AR, Cook ME, Adams EJ, Hildebrand WH, Gumperz JE. PLoS Biol. 2009;7:e1000228. doi: 10.1371/journal.pbio.1000228. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 240.Allende ML, Zhou D, Kalkofen DN, Benhamed S, Tuymetova G, Borowski C, Bendelac A, Proia RL. F A S E B J. 2008;22:307. doi: 10.1096/fj.07-9087com. [DOI] [PubMed] [Google Scholar]
- 241.Hwang SJ, Kim JH, Kim HY, Kim S, Chung DH. Lab Invest. 2010;90:9. doi: 10.1038/labinvest.2009.109. [DOI] [PubMed] [Google Scholar]
- 242.Graeler M, Goetzl EJ. F A S E B J. 2002;16:1874. doi: 10.1096/fj.02-0548com. [DOI] [PubMed] [Google Scholar]
- 243.Allende ML, Dreier JL, Mandala S, Proia RL. J Biol Chem. 2004;279:15396. doi: 10.1074/jbc.M314291200. [DOI] [PubMed] [Google Scholar]
- 244.Matloubian M, Lo CG, Cinamon G, Lesneski MJ, Xu Y, Brinkmann V, Allende ML, Proia RL, Cyster JG. Nature. 2004;427:355. doi: 10.1038/nature02284. [DOI] [PubMed] [Google Scholar]
- 245.Lo CG, Xu Y, Proia RL, Cyster JG. J Exp Med. 2005;201:291. doi: 10.1084/jem.20041509. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 246.Thangada S, Khanna KM, Blaho VA, Oo ML, Im DS, Guo C, Lefrancois L, Hla T. J Exp Med. 2010;207:1475. doi: 10.1084/jem.20091343. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 247.Pham TH, Okada T, Matloubian M, Lo CG, Cyster JG. Immunity. 2008;28:122. doi: 10.1016/j.immuni.2007.11.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 248.Yopp AC, Ochando JC, Mao M, Ledgerwood L, Ding Y, Bromberg JS. J Immunol. 2005;175:2913. doi: 10.4049/jimmunol.175.5.2913. [DOI] [PubMed] [Google Scholar]
- 249.Shiow LR, Rosen DB, Brdičková N, Xu Y, An J, Lanier LL, Cyster JG, Matloubian M. Nature. 2006;440:540. doi: 10.1038/nature04606. [DOI] [PubMed] [Google Scholar]
- 250.Grigorova IL, Panteleev M, Cyster JG. Proc Nat Acad Sci U S A. 2010;107:20447. doi: 10.1073/pnas.1009968107. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 251.Carlson CM, Endrizzi BT, Wu J, Ding X, Weinreich MA, Walsh ER, Wani MA, Lingrel JB, Hogquist KA, Jameson SC. Nature. 2006;442:299. doi: 10.1038/nature04882. [DOI] [PubMed] [Google Scholar]
- 252.Zachariah MA, Cyster JG. Science. 2010;328:1129. doi: 10.1126/science.1188222. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 253.Li Z, Zhang Y, Liu Z, Wu X, Zheng Y, Tao Z, Mao K, Wang J, Lin G, Tian L, Ji Y, Qin M, Sun S, Zhu X, Sun B. Nat Immunol. 2011;12:178. doi: 10.1038/ni.1983. [DOI] [PubMed] [Google Scholar]
- 254.Ledgerwood LG, Lal G, Zhang N, Garin A, Esses SJ, Ginhoux F, Merad M, Peche H, Lira SA, Ding Y, Yang Y, He X, Schuchman EH, Allende ML, Ochando JC, Bromberg JS. Nat Immunol. 2008;9:42. doi: 10.1038/ni1534. [DOI] [PubMed] [Google Scholar]
- 255.Brown MN, Fintushel SR, Lee MH, Jennrich S, Geherin SA, Hay JB, Butcher EC, Debes GF. J Immunol. 2010;185:4873. doi: 10.4049/jimmunol.1000676. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 256.Huang MC, Greig NH, Luo W, Tweedie D, Schwartz JB, Longo DL, Ferrucci L, Ershler WB, Goetzl EJ. Clin Immunol. 2010;138:201. doi: 10.1016/j.clim.2010.11.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 257.Grigorova IL, Schwab SR, Phan TG, Pham THM, Okada T, Cyster JG. Nat Immunol. 2009;10:58. doi: 10.1038/ni.1682. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 258.Kunisawa J, Kurashima Y, Higuchi M, Gohda M, Ishikawa I, Ogahara I, Kim N, Shimizu M, Kiyono H. J Exp Med. 2007;204:2335. doi: 10.1084/jem.20062446. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 259.Halin C, Scimone ML, Bonasio R, Gauguet JM, Mempel TR, Quackenbush E, Proia RL, Mandala S, von Andrian UH. Blood. 2005;106:1314. doi: 10.1182/blood-2004-09-3687. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 260.Gräler MH, Huang MC, Watson S, Goetzl EJ. J Immunol. 2005;174:1997. doi: 10.4049/jimmunol.174.4.1997. [DOI] [PubMed] [Google Scholar]
- 261.Dorsam G, Graeler MH, Seroogy C, Kong Y, Voice JK, Goetzl EJ. J Immunol. 2003;171:3500. doi: 10.4049/jimmunol.171.7.3500. [DOI] [PubMed] [Google Scholar]
- 262.Huang MC, Watson SR, Liao JJ, Goetzl EJ. J Immunol. 2007;178:6806. doi: 10.4049/jimmunol.178.11.6806. [DOI] [PubMed] [Google Scholar]
- 263.Liao JJ, Huang MC, Goetzl EJ. J Immunol. 2007;178:5425. doi: 10.4049/jimmunol.178.9.5425. [DOI] [PubMed] [Google Scholar]
- 264.Sawicka E, Dubois G, Jarai G, Edwards M, Thomas M, Nicholls A, Albert R, Newson C, Brinkmann V, Walker C. J Immunol. 2005;175:7973. doi: 10.4049/jimmunol.175.12.7973. [DOI] [PubMed] [Google Scholar]
- 265.Kim MG, Lee SY, Ko YS, Lee HY, Jo SK, Cho WY, Kim HK. Nephrol Dial Transplant. 2011;26:111. doi: 10.1093/ndt/gfq480. [DOI] [PubMed] [Google Scholar]
- 266.Liu G, Burns S, Huang G, Boyd K, Proia RL, Flavell RA, Chi H. Nat Immunol. 2009;10:769. doi: 10.1038/ni.1743. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 267.Liu G, Yang K, Burns S, Shrestha S, Chi H. Nat Immunol. 2010;11:1047. doi: 10.1038/ni.1939. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 268.Rubenfeld J, Guo J, Sookrung N, Chen R, Chaicumpa W, Casolaro V, Zhao Y, Natarajan V, Georas S. Am J Physiol Lung Cell Mol Physiol. 2006;290:66. doi: 10.1152/ajplung.00473.2004. [DOI] [PubMed] [Google Scholar]
- 269.Wang L, Knudsen E, Jin Y, Gessani S, Maghazachi AA. Cell Signal. 2004;16:991. doi: 10.1016/j.cellsig.2004.02.001. [DOI] [PubMed] [Google Scholar]
- 270.Goetzl EJ, Kong Y, Mei B. J Immunol. 1999;162:2049. [PubMed] [Google Scholar]
- 271.Goetzl EJ, Kong Y, Voice JK. J Immunol. 2000;164:4996. doi: 10.4049/jimmunol.164.10.4996. [DOI] [PubMed] [Google Scholar]
- 272.Zheng Y, Kong Y, Goetzl EJ. J Immunol. 2001;166:2317. doi: 10.4049/jimmunol.166.4.2317. [DOI] [PubMed] [Google Scholar]
- 273.Hu X, Haney N, Kropp D, Kabore AF, Johnston JB, Gibson SB. J Biol Chem. 2005;280:9498. doi: 10.1074/jbc.M410455200. [DOI] [PubMed] [Google Scholar]
- 274.Satoh Y, Ohkawa R, Nakamura K, Higashi K, Kaneko M, Yokota H, Aoki J, Arai H, Yuasa Y, Yatomi Y. Eur J Haematol. 2007;78:510. doi: 10.1111/j.1600-0609.2007.00849.x. [DOI] [PubMed] [Google Scholar]
- 275.Nam JH, Shin DH, Min JE, Ye SK, Jeon JH, Kim SJ. Mol Cells. 2010;29:85. doi: 10.1007/s10059-010-0020-4. [DOI] [PubMed] [Google Scholar]
- 276.Allende ML, Tuymetova G, Lee BG, Bonifacino E, Wu YP, Proia RL. J Exp Med. 2010;207:1113. doi: 10.1084/jem.20092210. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 277.Pereira JP, Xu Y, Cyster JG. PloS One. 2010;5:e9277. doi: 10.1371/journal.pone.0009277. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 278.Cinamon G, Matloubian M, Lesneski MJ, Xu Y, Low C, Lu T, Proia RL, Cyster JG. Nat Immunol. 2004;5:713. doi: 10.1038/ni1083. [DOI] [PubMed] [Google Scholar]
- 279.Girkontaite I, Sakk V, Wagner M, Borggrefe T, Tedford K, Chun J, Fischer KD. J Exp Med. 2004;200:1491. doi: 10.1084/jem.20041483. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 280.Kabashima K, Haynes NM, Xu Y, Nutt SL, Allende ML, Proia RL, Cyster JG. J Exp Med. 2006;203:2683. doi: 10.1084/jem.20061289. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 281.Kunisawa J, Kurashima Y, Gohda M, Higuchi M, Ishikawa I, Miura F, Ogahara I, Kiyono H. Blood. 2007;109:3749. doi: 10.1182/blood-2006-08-041582. [DOI] [PubMed] [Google Scholar]
- 282.Gohda M, Kunisawa J, Miura F, Kagiyama Y, Kurashima Y, Higuchi M, Ishikawa I, Ogahara I, Kiyono H. J Immunol. 2008;180:5335. doi: 10.4049/jimmunol.180.8.5335. [DOI] [PubMed] [Google Scholar]
- 283.Massberg S, Schaerli P, Knezevic-Maramica I, Köllnberger M, Tubo N, Moseman EA, Huff IV, Junt T, Wagers AJ, Mazo IB, von Andrian UH. Cell. 2007;131:994. doi: 10.1016/j.cell.2007.09.047. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 284.Ratajczak MZ, Lee H, Wysoczynski M, Wan W, Marlicz W, Laughlin MJ, Kucia M, Janowska-Wieczorek A, Ratajczak J. Leukemia. 2010;24:976. doi: 10.1038/leu.2010.53. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 285.Kimura T, Boehmler AM, Seitz G, Kuçi S, Wiesner T, Brinkmann V, Kanz L, Möhle R. Blood. 2004;103:4478. doi: 10.1182/blood-2003-03-0875. [DOI] [PubMed] [Google Scholar]
- 286.Walter DH, Rochwalsky U, Reinhold J, Seeger F, Aicher A, Urbich C, Spyridopoulos I, Chun J, Brinkmann V, Keul P, Levkau B, Zeiher AM, Dimmeler S, Haendeler J. Arterioscler Thromb Vasc Biol. 2007;27:275. doi: 10.1161/01.ATV.0000254669.12675.70. [DOI] [PubMed] [Google Scholar]
- 287.Ishii M, Egen JG, Klauschen F, Meier-Schellersheim M, Saeki Y, Vacher J, Proia RL, Germain RN. Nature. 2009;458:524. doi: 10.1038/nature07713. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 288.Ishii M, Kikuta J, Shimazu Y, Meier-Schellersheim M, Germain RN. The J Exp Med. 2010;207:2793. doi: 10.1084/jem.20101474. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 289.Yanai N, Matsui N, Furusawa T, Okubo T, Obinata M. Blood. 2000;96:139. [PubMed] [Google Scholar]
- 290.Liu YB, Kharode Y, Bodine PVN, Yaworsky PJ, Robinson JA, Billiard J. J Cell Biochem. 2010;109:794. doi: 10.1002/jcb.22471. [DOI] [PubMed] [Google Scholar]
- 291.Gennero I, Laurencin-Dalicieux S, Conte-Auriol F, Briand-Mésange F, Laurencin D, Rue J, Beton N, Malet N, Mus M, Tokumura A, Bourin P, Vico L, Brunel G, Oreffo ROC, Chun J, Salles JP. Bone. 2011;329:283. doi: 10.1016/j.bone.2011.04.018. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 292.Lin ME, Herr DR, Chun J. Prostaglandins Other Lipid Med. 2010;91:130. doi: 10.1016/j.prostaglandins.2009.02.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 293.Dubin AE, Herr DR, Chun J. J Neurosci. 2010;30:7300. doi: 10.1523/JNEUROSCI.6151-09.2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 294.Fukushima N, Kimura Y, Chun J. Proc Nat Acad Sci U S A. 1998;95:6151. doi: 10.1073/pnas.95.11.6151. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 295.Fukushima N, Weiner JA, Chun J. Develop Biol. 2000;228:6. doi: 10.1006/dbio.2000.9930. [DOI] [PubMed] [Google Scholar]
- 296.Estivill-Torrús G, Llebrez-Zayas P, Matas-Rico E, Santín L, Pedraza C, De Diego I, Del Arco I, Fernández-Llebrez P, Chun J, De Fonseca FR. Cereb Cortex. 2008;18:938. doi: 10.1093/cercor/bhm132. [DOI] [PubMed] [Google Scholar]
- 297.Hurst JH, Mumaw J, Machacek DW, Sturkie C, Callihan P, Stice SL, Hooks SB. B M C Neurosci. 2008;9:118. doi: 10.1186/1471-2202-9-118. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 298.Kingsbury MA, Rehen SK, Contos JJA, Higgins CM, Chun J. Nat Neurosci. 2003;6:1292–1299. doi: 10.1038/nn1157. [DOI] [PubMed] [Google Scholar]
- 299.Fukushima N, Shano S, Moriyama R, Chun J. Neurochem Internat. 2007;50:302–307. doi: 10.1016/j.neuint.2006.09.008. [DOI] [PubMed] [Google Scholar]
- 300.Yanagida K, Ishii S, Hamano F, Noguchi K, Shimizu T. J Biol Chem. 2007;282:5814. doi: 10.1074/jbc.M610767200. [DOI] [PubMed] [Google Scholar]
- 301.Sayas CL, Ariaens A, Ponsioen B, Moolenaar WH. Mol Biol Cell. 2006;17:1834. doi: 10.1091/mbc.E05-07-0688. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 302.Sun Y, Kim NH, Yang H, Kim SH, Huh SO. Mol Cells. 2011;31:483. doi: 10.1007/s10059-011-1036-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 303.Harada J, Foley M, Moskowitz MA, Waeber C. J Neurochem. 2004;88:1026. doi: 10.1046/j.1471-4159.2003.02219.x. [DOI] [PubMed] [Google Scholar]
- 304.Kimura A, Ohmori T, Ohkawa R, Madoiwa S, Mimuro J, Murakami T, Kobayashi E, Hoshino Y, Yatomi Y, Sakata Y. Stem cells (Dayton, Ohio) 2007;25:115. doi: 10.1634/stemcells.2006-0223. [DOI] [PubMed] [Google Scholar]
- 305.Giaume C, Koulakoff A, Roux L, Holcman D, Rouach N. Nat Rev Neurosci. 2010;11:87. doi: 10.1038/nrn2757. [DOI] [PubMed] [Google Scholar]
- 306.Shano S, Moriyama R, Chun J, Fukushima N. Neurochem Internat. 2008;52:216. doi: 10.1016/j.neuint.2007.07.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 307.Sorensen SD, Nicole O, Peavy RD, Montoya LM, Lee CJ, Murphy TJ, Traynelis SF, Hepler JR. Mol Pharmacol. 2003;64:1199. doi: 10.1124/mol.64.5.1199. [DOI] [PubMed] [Google Scholar]
- 308.Bassi R, Anelli V, Giussani P, Tettamanti G, Viani P, Riboni L. Glia. 2006;53:621. doi: 10.1002/glia.20324. [DOI] [PubMed] [Google Scholar]
- 309.Pébay A, Toutant M, Prémont J, Calvo CF, Venance L, Cordier J, Glowinski J, Tencé M. Eur J Neurosci. 2001;13:2067. [PubMed] [Google Scholar]
- 310.Rao TS, Lariosa-Willingham KD, Lin FF, Yu N, Tham CS, Chun J, Webb M. Int J Dev Neurosci. 2004;22:131. doi: 10.1016/j.ijdevneu.2004.03.005. [DOI] [PubMed] [Google Scholar]
- 311.Spohr TCdSe, Choi JW, Gardell SE, Herr DR, Rehen SK, Gomes FCA, Chun J. J Biol Chem. 2008;283:7470. doi: 10.1074/jbc.M707758200. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 312.Rouach N, Pébay A, Même W, Cordier J, Ezan P, Etienne E, Giaume C, Tencé M. Eur J Neurosci. 2006;23:1453. doi: 10.1111/j.1460-9568.2006.04671.x. [DOI] [PubMed] [Google Scholar]
- 313.Choi JW, Gardell SE, Herr DR, Rivera R, Lee CW, Noguchi K, Teo ST, Yung YC, Lu M, Kennedy G, Chun J. Proc Nat Acad Sci U S A. 2010;108:751. doi: 10.1073/pnas.1014154108. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 314.Ginhoux F, Greter M, Leboeuf M, Nandi S, See P, Gokhan S, Mehler MF, Conway SJ, Ng LG, Stanley ER, Samokhvalov IM, Merad M. Science. 2010;330:841. doi: 10.1126/science.1194637. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 315.Moller T, Contos JJ, Musante DB, Chun J, Ransom BR. J Biol Chem. 2001;276:25946. doi: 10.1074/jbc.M102691200. [DOI] [PubMed] [Google Scholar]
- 316.Bernhart E, Kollroser M, Rechberger G, Reicher H, Heinemann A, Schratl P, Hallström S, Wintersperger A, Nusshold C, DeVaney T, Zorn-Pauly K, Malli R, Graier W, Malle E, Sattler W. Proteomics. 2010;10:141. doi: 10.1002/pmic.200900195. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 317.Tham CS, Lin FF, Rao TS, Yu N, Webb M. Int J Dev Neurosci. 2003;21:431. doi: 10.1016/j.ijdevneu.2003.09.003. [DOI] [PubMed] [Google Scholar]
- 318.Fujita R, Ma Y, Ueda H. J Neurochem. 2008;107:152. doi: 10.1111/j.1471-4159.2008.05599.x. [DOI] [PubMed] [Google Scholar]
- 319.Durafourt BA, Lambert C, Johnson TA, Blain M, Bar-Or A, Antel JP. J Neuroimmunol. 2011;230:10. doi: 10.1016/j.jneuroim.2010.08.006. [DOI] [PubMed] [Google Scholar]
- 320.Miron VE, Ludwin SK, Darlington PJ, Jarjour AA, Soliven B, Kennedy TE, Antel JP. Am J Pathol. 2010;176:2682. doi: 10.2353/ajpath.2010.091234. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 321.Jackman N, Ishii A, Bansal R. Physiology (Bethesda, Md) 2009;24:290. doi: 10.1152/physiol.00016.2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 322.Weiner JA, Hecht JH, Chun J. J Comp Neurol. 1998;398:587. [PubMed] [Google Scholar]
- 323.Yu N, Lariosa-Willingham KD, Lin FF, Webb M, Rao TS. Glia. 2004;45:17. doi: 10.1002/glia.10297. [DOI] [PubMed] [Google Scholar]
- 324.Nogaroli L, Yuelling LM, Dennis J, Gorse K, Payne SG, Fuss B. Neurochem Res. 2009;34:182. doi: 10.1007/s11064-008-9772-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 325.Im DS, Clemens J, Macdonald TL, Lynch KR. Biochemistry. 2001;40:14053. doi: 10.1021/bi011606i. [DOI] [PubMed] [Google Scholar]
- 326.Novgorodov AS, El-Alwani M, Bielawski J, Obeid LM, Gudz TI. F A S E B J. 2007;21:1503. doi: 10.1096/fj.06-7420com. [DOI] [PubMed] [Google Scholar]
- 327.Jaillard C, Harrison S, Stankoff B, Aigrot MS, Calver AR, Duddy G, Walsh FS, Pangalos MN, Arimura N, Kaibuchi K, Zalc B, Lubetzki C. J Neurosci. 2005;25:1459. doi: 10.1523/JNEUROSCI.4645-04.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 328.Jung CG, Kim HJ, Miron VE, Cook S, Kennedy TE, Foster CA, Antel JP, Soliven B. Glia. 2007;55:1656. doi: 10.1002/glia.20576. [DOI] [PubMed] [Google Scholar]
- 329.Weiner JA, Chun J. Proc Nat Acad Sci U S A. 1999;96:5233. doi: 10.1073/pnas.96.9.5233. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 330.Weiner JA, Fukushima N, Contos JJ, Scherer SS, Chun J. J Neurosci. 2001;21:7069. doi: 10.1523/JNEUROSCI.21-18-07069.2001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 331.Barber SC, Mellor H, Gampel A, Scolding NJ. Eur J Neurosci. 2004;19:3142. doi: 10.1111/j.0953-816X.2004.03424.x. [DOI] [PubMed] [Google Scholar]
- 332.Yuan Xb, Jin M, Xu X, Song Yq, Wu Cp, Poo Mm, Duan S. Nat Cell Biol. 2003;5:38. doi: 10.1038/ncb895. [DOI] [PubMed] [Google Scholar]
- 333.Goldshmit Y, Munro K, Leong SY, Pébay A, Turnley AM. Cell Tissue Res. 2010;341:23. doi: 10.1007/s00441-010-0977-5. [DOI] [PubMed] [Google Scholar]
- 334.Inoue M, Rashid MH, Fujita R, Contos JJA, Chun J, Ueda H. Nat Med. 2004;10:712. doi: 10.1038/nm1060. [DOI] [PubMed] [Google Scholar]
- 335.Inoue M, Yamaguchi A, Kawakami M, Chun J, Ueda H. Molec Pain. 2006;2:25. doi: 10.1186/1744-8069-2-25. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 336.Ahn DK, Lee SY, Han SR, Ju JS, Yang GY, Lee MK, Youn DH, Bae YC. Pain. 2009;146:114. doi: 10.1016/j.pain.2009.07.012. [DOI] [PubMed] [Google Scholar]
- 337.Nagai J, Uchida H, Matsushita Y, yano R, Ueda M, Niwa M, Aoki J, Chun J, Ueda H. Molec Pain. 2010;6:78. doi: 10.1186/1744-8069-6-78. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 338.Castilla-Ortega E, Sánchez-López J, Hoyo-Becerra C, Matas-Rico E, Zambrana-Infantes E, Chun J, De Fonseca FR, Pedraza C, Estivill-Torrús G, Santin LJ. Neurobiol Learn Mem. 2010;94:73. doi: 10.1016/j.nlm.2010.04.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 339.Zhang YH, Fehrenbacher JC, Vasko MR, Nicol GD. J Neurophysiol. 2006;96:1042. doi: 10.1152/jn.00120.2006. [DOI] [PubMed] [Google Scholar]
- 340.Chi XX, Nicol GD. J Neurophysiol. 2010;104:2741. doi: 10.1152/jn.00709.2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 341.Strochlic L, Dwivedy A, van Horck FPG, Falk J, Holt CE. Development (Cambridge, England) 2008;135:333. doi: 10.1242/dev.009563. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 342.Crousillac S, Colonna J, McMains E, Dewey JS, Gleason E. J Neurophysiol. 2009;102:3295. doi: 10.1152/jn.00119.2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 343.Forrest M, Sun SY, Hajdu R, Bergstrom J, Card D, Doherty G, Hale J, Keohane C, Meyers C, Milligan J, Mills S, Nomura N, Rosen H, Rosenbach M, Shei GJ, Singer II, Tian M, West S, White V, Xie J, Proia RL, Mandala S. J Pharmacol Exp Ther. 2004;309:758. doi: 10.1124/jpet.103.062828. [DOI] [PubMed] [Google Scholar]
- 344.McCain DF, Wu L, Nickel P, Kassack MU, Kreimeyer A, Gagliardi A, Collins DC, Zhang ZY. J Biol Chem. 2004;279:14713. doi: 10.1074/jbc.M312488200. [DOI] [PubMed] [Google Scholar]
- 345.Tokumura A, Kanaya Y, Miyake M, Yamano S, Irahara M, Fukuzawa K. Biol Reprod. 2002;67:1386. doi: 10.1095/biolreprod.102.004051. [DOI] [PubMed] [Google Scholar]
- 346.Wang H, Dey SK. Nat Rev Genetics. 2006;7:185. doi: 10.1038/nrg1808. [DOI] [PubMed] [Google Scholar]
- 347.Kamińska K, Wasielak M, Bogacka I, Blitek M, Bogacki M. Prostaglandins Other Lipid Med. 2008;85:26. doi: 10.1016/j.prostaglandins.2007.10.001. [DOI] [PubMed] [Google Scholar]
- 348.Noguchi K, Ishii S, Shimizu T. J Biol Chem. 2003;278:25600. doi: 10.1074/jbc.M302648200. [DOI] [PubMed] [Google Scholar]
- 349.Chen SU, Lee H, Chang DY, Chou CH, Chang CY, Chao KH, Lin CW, Yang YS. Endocrinology. 2008;149:5888. doi: 10.1210/en.2008-0314. [DOI] [PubMed] [Google Scholar]
- 350.Chen SU, Chou CH, Chao KH, Lee H, Lin CW, Lu HF, Yang YS. Endocrinology. 2010;151:369. doi: 10.1210/en.2009-0779. [DOI] [PubMed] [Google Scholar]
- 351.Dunlap KA, Kwak Hi, Burghardt RC, Bazer FW, Magness RR, Johnson GA, Bayless KJ. Biol Reprod. 2010;82:876. doi: 10.1095/biolreprod.109.081604. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 352.Skaznik-Wikiel ME, Kaneko-Tarui T, Kashiwagi A, Pru JK. Biol Reprod. 2006;74:569. doi: 10.1095/biolreprod.105.046714. [DOI] [PubMed] [Google Scholar]
- 353.Mizugishi K, Li C, Olivera A, Bielawski J, Bielawska A, Deng CX, Proia RL. J Clin Invest. 2007;117:2993. doi: 10.1172/JCI30674. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 354.Yamamoto Y, Olson DM, van Bennekom M, Brindley DN, Hemmings DG. Biol Reprod. 2010;82:628. doi: 10.1095/biolreprod.109.081497. [DOI] [PubMed] [Google Scholar]
- 355.Ye X, Skinner MK, Kennedy G, Chun J. Biol Reprod. 2008;79:328. doi: 10.1095/biolreprod.108.068783. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 356.Suomalainen L, Hakala JK, Pentikäinen V, Otala M, Erkkilä K, Pentikäinen MO, Dunkel L. J Clin Endocrinol Metab. 2003;88:5572. doi: 10.1210/jc.2003-030776. [DOI] [PubMed] [Google Scholar]
- 357.Matsumoto K, Banno Y, Murate T, Akao Y, Nozawa Y. J Histochem Cytochem. 2005;53:243. doi: 10.1369/jhc.4B6507.2005. [DOI] [PubMed] [Google Scholar]
- 358.Chae SS, Proia RL, Hla T. Prostaglandins Other Lipid Med. 2004;73:141. doi: 10.1016/j.prostaglandins.2004.01.006. [DOI] [PubMed] [Google Scholar]
- 359.di Villa Bianca RdaE, Sorrentino R, Sorrentino R, Imbimbo C, Palmieri A, Fusco F, Maggi M, De Palma R, Cirino G, Mirone V. J Pharmacol Exp Ther. 2006;316:703. doi: 10.1124/jpet.105.093419. [DOI] [PubMed] [Google Scholar]
- 360.Bosier B, Hermans E. Trends Pharmacol Sci. 2007;28:438. doi: 10.1016/j.tips.2007.06.001. [DOI] [PubMed] [Google Scholar]
- 361.Evans JF, Hutchinson JH. Nat Chem Biol. 2010;6:476. doi: 10.1038/nchembio.394. [DOI] [PubMed] [Google Scholar]
- 362.Albers HMHG, van Meeteren LA, Egan DA, van Tilburg EW, Moolenaar WH, Ovaa H. J Med Chem. 2010;53:4958. doi: 10.1021/jm1005012. [DOI] [PubMed] [Google Scholar]
- 363.East JE, Kennedy AJ, Tomsig JL, De Leon AR, Lynch KR, Macdonald TL. Bioorg Med Chem Lett. 2010;20:7132. doi: 10.1016/j.bmcl.2010.09.030. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 364.Gierse J, Thorarensen A, Beltey K, Bradshaw-Pierce E, Cortes-Burgos L, Hall T, Johnston A, Murphy M, Nemirovskiy O, Ogawa S, Pegg L, Pelc M, Prinsen M, Schnute M, Wendling J, Wene S, Weinberg R, Wittwer A, Zweifel B, Masferrer J. J Pharmacol Exp Ther. 2010;334:310. doi: 10.1124/jpet.110.165845. [DOI] [PubMed] [Google Scholar]
- 365.Albers HMHG, Dong A, van Meeteren LA, Egan DA, Sunkara M, van Tilburg EW, Schuurman K, van Tellingen O, Morris AJ, Smyth SS, Moolenaar WH, Ovaa H. Proc Nat Acad Sci U S A. 2010;107:7257. doi: 10.1073/pnas.1001529107. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 366.Ferry G, Moulharat N, Pradere JP, Desos P, Try A, Genton A, Giganti A, Beucher-Gaudin M, Lonchampt M, Bertrand M, Saulnier-Blache JS, Tucker GC, Cordi A, Boutin JA. J Pharm Exp Therap. 2008;327:809. doi: 10.1124/jpet.108.141911. [DOI] [PubMed] [Google Scholar]
- 367.Gupte R, Patil R, Liu J, Wang Y, Lee SC, Fujiwara Y, Fells J, Bolen AL, Emmons-Thompson K, Yates CR, Siddam A, Panupinthu N, Pham TCT, Baker DL, Parrill AL, Mills GB, Tigyi G, Miller DD. Chem Med Chem. 2011;6:922. doi: 10.1002/cmdc.201000425. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 368.Castelino FV, Seiders J, Bain G, Brooks SF, King CD, Swaney JS, Lorrain DS, Chun J, Luster AD, Tager AM. Arth Rheum. 2011;63:1405. doi: 10.1002/art.30262. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 369.Swaney JS, Chapman C, Correa LD, Stebbins KJ, Broadhead AR, Bain G, Santini AM, Darlington J, King CD, Baccei CS, Lee C, Parr TA, Roppe JR, Seiders TJ, Ziff J, Prasit P, Hutchinson JH, Evans JF, Lorrain DS. J Pharm Exp Therap. 2011;336:693. doi: 10.1124/jpet.110.175901. [DOI] [PubMed] [Google Scholar]
- 370.Amira Pharmaceuticals Receives Orphan Drug Status for a Novel LPA1 Antagonist, AM152, for the Treatment of Idiopathic Pulmonary Fibrosis. 2011 http://www.amirapharm.com/articles/AM152 Orphan Drug.html.
- 371.Swaney JS, Chapman C, Correa LD, Stebbins KJ, Bundey RA, Prodanovich PC, Fagan P, Baccei CS, Santini AM, Hutchinson JH, Seiders TJ, Parr TA, Prasit P, Evans JF, Lorrain DS. Br J Pharmacol. 2010;160:1699. doi: 10.1111/j.1476-5381.2010.00828.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 372.Ohta H, Sato K, Murata N, Damirin A, Malchinkhuu E, Kon J, Kimura T, Tobo M, Yamazaki Y, Watanabe T, Yagi M, Sato M, Suzuki R, Murooka H, Sakai T, Nishitoba T, Im DS, Nochi H, Tamoto K, Tomura H, Okajima F. Mol Pharmacol. 2003;64:994. doi: 10.1124/mol.64.4.994. [DOI] [PubMed] [Google Scholar]
- 373.Snider AJ, Zhang Z, Xie Y, Meier KE. Am J Physiol Cell Physiol. 2010;298:C163. doi: 10.1152/ajpcell.00001.2009. [DOI] [PubMed] [Google Scholar]
- 374.Park SY, Jeong KJ, Panupinthu N, Yu S, Lee J, Han JW, Kim JM, Lee JS, Kang J, Park CG, Mills GB, Lee HY. Oncogene. 2010;30:1351. doi: 10.1038/onc.2010.517. [DOI] [PubMed] [Google Scholar]
- 375.Subramanian P, Karshovska E, Reinhard P, Megens RTA, Zhou Z, Akhtar S, Schumann U, Li X, van Zandvoort M, Ludin C, Weber C, Schober A. Circ Res. 2010;107:96. doi: 10.1161/CIRCRESAHA.109.212647. [DOI] [PubMed] [Google Scholar]
- 376.Heise CE, Santos WL, Schreihofer AM, Heasley BH, Mukhin YV, Macdonald TL, Lynch KR. Mol Pharmacol. 2001;60:1173. doi: 10.1124/mol.60.6.1173. [DOI] [PubMed] [Google Scholar]
- 377.Yamada T, Sato K, Komachi M, Malchinkhuu E, Tobo M, Kimura T, Kuwabara A, Yanagita Y, Ikeya T, Tanahashi Y, Ogawa T, Ohwada S, Morishita Y, Ohta H, Im DS, Tamoto K, Tomura H, Okajima F. J Biol Chem. 2004;279:6595. doi: 10.1074/jbc.M308133200. [DOI] [PubMed] [Google Scholar]
- 378.Hasegawa Y, Erickson JR, Goddard GJ, Yu S, Liu S, Cheng KW, Eder A, Bandoh K, Aoki J, Jarosz R, Schrier AD, Lynch KR, Mills GB, Fang X. J Biol Chem. 2003;278:11962. doi: 10.1074/jbc.M209168200. [DOI] [PubMed] [Google Scholar]
- 379.Okusa MD, Ye H, Huang L, Sigismund L, Macdonald T, Lynch KR. Am J Physiol Renal Physiol. 2003;285:F565. doi: 10.1152/ajprenal.00023.2003. [DOI] [PubMed] [Google Scholar]
- 380.Cohen JA, Chun J. Ann Neurol. 2011;69:759. doi: 10.1002/ana.22426. [DOI] [PubMed] [Google Scholar]
- 381.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. Science. 2002;296:346. doi: 10.1126/science.1070238. [DOI] [PubMed] [Google Scholar]
- 382.Brinkmann V, Davis MD, Heise CE, Albert R, Cottens S, Hof R, Bruns C, Prieschl E, Baumruker T, Hiestand P, Foster CA, Zollinger M, Lynch KR. J Biol Chem. 2002;277:21453. doi: 10.1074/jbc.C200176200. [DOI] [PubMed] [Google Scholar]
- 383.Billich A, Bornancin F, Dévay P, Mechtcheriakova D, Urtz N, Baumruker T. J Biol Chem. 2003;278:47408. doi: 10.1074/jbc.M307687200. [DOI] [PubMed] [Google Scholar]
- 384.Oo ML, Thangada S, Wu MT, Liu CH, Macdonald TL, Lynch KR, Lin CY, Hla T. J Biol Chem. 2007;282:9082. doi: 10.1074/jbc.M610318200. [DOI] [PubMed] [Google Scholar]
- 385.Budde K, Schmouder RL, Brunkhorst R, Nashan B, Lücker PW, Mayer T, Choudhury S, Skerjanec A, Kraus G, Neumayer HH. J Am Soc Nephrol. 2002;13:1073. doi: 10.1681/ASN.V1341073. [DOI] [PubMed] [Google Scholar]
- 386.Salvadori M, Budde K, Charpentier B, Klempnauer J, Nashan B, Pallardo LM, Eris J, Schena FP, Eisenberger U, Rostaing L, Hmissi A, Aradhye S, Group FS. Am J Transplant. 2006;6:2912. doi: 10.1111/j.1600-6143.2006.01552.x. [DOI] [PubMed] [Google Scholar]
- 387.Tedesco-Silva H, Szakaly P, Shoker A, Sommerer C, Yoshimura N, Schena F, Cremer M, Hmissi A, Mayer H, Lang P. Transplantation. 2007;84:885. doi: 10.1097/01.tp.0000281385.26500.3b. [DOI] [PubMed] [Google Scholar]
- 388.Webb M, Tham CS, Lin FF, Lariosa-Willingham K, Yu N, Hale J, Mandala S, Chun J, Rao TS. J Neuroimmunol. 2004;153:108. doi: 10.1016/j.jneuroim.2004.04.015. [DOI] [PubMed] [Google Scholar]
- 389.Foster CA, Howard LM, Schweitzer A, Persohn E, Hiestand PC, Balatoni B, Reuschel R, Beerli C, Schwartz M, Billich A. J Pharmacol Exp Ther. 2007;323:469. doi: 10.1124/jpet.107.127183. [DOI] [PubMed] [Google Scholar]
- 390.Van Doorn R, Van Horssen J, Verzijl D, Witte M, Ronken E, Van Het Hof B, Lakeman K, Dijkstra CD, Van Der Valk P, Reijerkerk A, Alewijnse AE, Peters SLM, De Vries HE. Glia. 2010;58:1465. doi: 10.1002/glia.21021. [DOI] [PubMed] [Google Scholar]
- 391.Soliven B, Miron V, Chun J. Neurology. 2011;76:S9. doi: 10.1212/WNL.0b013e31820d9507. [DOI] [PubMed] [Google Scholar]
- 392.Hla T, Brinkmann V. Neurology. 2011;76:S3. doi: 10.1212/WNL.0b013e31820d5ec1. [DOI] [PubMed] [Google Scholar]
- 393.Noguchi K, Chun J. Crit Rev Biochem Mol Biol. 2011;46:2. doi: 10.3109/10409238.2010.522975. [DOI] [PubMed] [Google Scholar]
- 394.Morris MA, Mcduffie M, Nadler JL, Ley K. Autoimmunity. 2011;44:115. doi: 10.3109/08916934.2010.499885. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 395.Kendall MR, Hupfeld CJ. Diabetes Obesity Metab. 2008;10:802. doi: 10.1111/j.1463-1326.2008.00910.x. [DOI] [PubMed] [Google Scholar]
- 396.Commodaro AG, Peron JP, Lopes CT, Arslanian C, Belfort R, Rizzo LV, Bueno V. Invest Opthalmol Vis Sci. 2010;51:2568. doi: 10.1167/iovs.09-4769. [DOI] [PubMed] [Google Scholar]
- 397.Pan S, Mi Y, Pally C, Beerli C, Chen A, Guerini D, Hinterding K, Nuesslein-Hildesheim B, Tuntland T, Lefebvre S. Chem Biol. 2006;13:1227. doi: 10.1016/j.chembiol.2006.09.017. [DOI] [PubMed] [Google Scholar]
- 398.Oo ML, Chang SH, Thangada S, Wu MT, Rezaul K, Blaho V, Hwang SI, Han DK, Hla T. J Clin Invest. 2011;121:2290. doi: 10.1172/JCI45403. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 399.Zhang ZY, Zhang Z, Zug C, Nuesslein-Hildesheim B, Leppert D, Schluesener HJ. J Neuroimmunol. 2009;216:59. doi: 10.1016/j.jneuroim.2009.09.010. [DOI] [PubMed] [Google Scholar]
- 400.Sanna MG, Liao J, Jo E, Alfonso C, Ahn MY, Peterson MS, Webb B, Lefebvre S, Chun J, Gray N, Rosen H. J Biol Chem. 2004;279:13839. doi: 10.1074/jbc.M311743200. [DOI] [PubMed] [Google Scholar]
- 401.Jo E, Sanna MG, Gonzalez-Cabrera PJ, Thangada S, Tigyi G, Osborne DA, Hla T, Parrill AL, Rosen H. Chem Biol. 2005;12:703. doi: 10.1016/j.chembiol.2005.04.019. [DOI] [PubMed] [Google Scholar]
- 402.Wei SH, Rosen H, Matheu MP, Sanna MG, Wang SK, Jo E, Wong CH, Parker I, Cahalan MD. Nat Immunol. 2005;6:1228. doi: 10.1038/ni1269. [DOI] [PubMed] [Google Scholar]
- 403.Skoura A, Hla T. Cardiovasc Res. 2009;82:221. doi: 10.1093/cvr/cvp088. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 404.Green JA, Suzuki K, Cho B, Willison LD, Palmer D, Allen CDC, Schmidt TH, Xu Y, Proia RL, Coughlin SR, Cyster JG. Nat Immunol. 2011;12:672. doi: 10.1038/ni.2047. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 405.Salomone S, Waeber C. Front Pharmacol. 2011;2:9. doi: 10.3389/fphar.2011.00009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 406.Long JS, Fujiwara Y, Edwards J, Tannahill CL, Tigyi G, Pyne S, Pyne NJ. J Biol Chem. 2010;285:35957. doi: 10.1074/jbc.M110.117945. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 407.Xia P, Gamble JR, Rye KA, Wang L, Hii CS, Cockerill P, Khew-Goodall Y, Bert AG, Barter PJ, Vadas MA. Proc Nat Acad Sci U S A. 1998;95:14196. doi: 10.1073/pnas.95.24.14196. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 408.Xia P, Wang L, Gamble JR, Vadas MA. J Biol Chem. 1999;274:34499. doi: 10.1074/jbc.274.48.34499. [DOI] [PubMed] [Google Scholar]
- 409.Olivera A, Buckley NE, Spiegel S. J Biol Chem. 1992;267:26121. [PubMed] [Google Scholar]
- 410.Olivera A, Kohama T, Edsall L, Nava V, Cuvillier O, Poulton S, Spiegel S. J Cell Biol. 1999;147:545. doi: 10.1083/jcb.147.3.545. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 411.Hannun YA, Obeid LM. J Biol Chem. 2011;286:27855. doi: 10.1074/jbc.R111.254359. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 412.Watson SP, McConnell RT, Lapetina EG. Biochem J. 1985;232:61. doi: 10.1042/bj2320061. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 413.Tokumura A, Fukuzawa K, Tsukatani H. Lipids. 1978;13:572. doi: 10.1007/BF02533598. [DOI] [PubMed] [Google Scholar]
- 414.Zondag GC, Postma FR, Etten IV, Verlaan I, Moolenaar WH. Biochem J. 1998;330(Pt 2):605. doi: 10.1042/bj3300605. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 415.Tolan D, Conway AM, Rakhit S, Pyne N, Pyne S. Cel Signal. 1999;11:349. doi: 10.1016/s0898-6568(99)00005-4. [DOI] [PubMed] [Google Scholar]
- 416.Igarashi Y, Yatomi Y. Acta Biochim Pol. 1998;45:299. [PubMed] [Google Scholar]
- 417.Chen JH, Riazy M, Wang SW, Dai JM, Duronio V, Steinbrecher UP. J Lipid Res. 2010;51:991. doi: 10.1194/jlr.M000398. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 418.Meyer Zu Heringdorf D, Liliom K, Schaefer M, Danneberg K, Jaggar JH, Tigyi G, Jakobs KH. F E B S Lett. 2003;554:443. doi: 10.1016/s0014-5793(03)01219-5. [DOI] [PubMed] [Google Scholar]
- 419.Park Hopson K, Truelove J, Chun J, Wang Y, Waeber C. Am J Physiol Cell Physiol. 2011;300:C919. doi: 10.1152/ajpcell.00350.2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 420.Berdyshev EV, Gorshkova I, Usatyuk P, Kalari S, Zhao Y, Pyne NJ, Pyne S, Sabbadini RA, Garcia JGN, Natarajan V. PloS One. 2011;6:e16571. doi: 10.1371/journal.pone.0016571. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 421.Hait NC, Allegood J, Maceyka M, Strub GM, Harikumar KB, Singh SK, Luo C, Marmorstein R, Kordula T, Milstien S, Spiegel S. Science. 2009;325:1254. doi: 10.1126/science.1176709. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 422.Alvarez SE, Harikumar KB, Hait NC, Allegood J, Strub GM, Kim EY, Maceyka M, Jiang H, Luo C, Kordula T, Milstien S, Spiegel S. Nature. 2010;465:1084. doi: 10.1038/nature09128. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 423.Yonamine I, Bamba T, Nirala NK, Jesmin N, Kosakowska-Cholody T, Nagashima K, Fukusaki E, Acharya JK, Acharya U. J Cell Biol. 2011;192:557. doi: 10.1083/jcb.201004098. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 424.Strub GM, Paillard M, Liang J, Gomez L, Allegood JC, Hait NC, Maceyka M, Price MM, Chen Q, Simpson DC, Kordula T, Milstien S, Lesnefsky EJ, Spiegel S. F A S E B J. 2011;25:600. doi: 10.1096/fj.10-167502. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 425.McIntyre TM, Pontsler AV, Silva AR, St Hilaire A, Xu Y, Hinshaw JC, Zimmerman GA, Hama K, Aoki J, Arai H, Prestwich GD. Proc Nat Acad Sci U S A. 2003;100:131. doi: 10.1073/pnas.0135855100. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 426.Stapleton CM, Mashek DG, Wang S, Nagle CA, Cline GW, Thuillier P, Leesnitzer LM, Li LO, Stimmel JB, Shulman GI, Coleman RA. PloS One. 2011;6:e18932. doi: 10.1371/journal.pone.0018932. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 427.Cheng Y, Makarova N, Tsukahara R, Guo H, Shuyu E, Farrar P, Balazs L, Zhang C, Tigyi G. Cel Signal. 2009;21:1874. doi: 10.1016/j.cellsig.2009.08.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 428.Prieschl EE, Csonga R, Novotny V, Kikuchi GE, Baumruker T. J Exp Med. 1999;190:1. doi: 10.1084/jem.190.1.1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 429.Pushparaj PN, Manikandan J, Tay HK, H'ng SC, Kumar SD, Pfeilschifter J, Huwiler A, Melendez AJ. J Immunol. 2009;183:221. doi: 10.4049/jimmunol.0803430. [DOI] [PubMed] [Google Scholar]
- 430.Simon MF, Daviaud D, Pradère JP, Grés S, Guigné C, Wabitsch M, Chun J, Valet P, Saulnier-Blache JS. J Biol Chem. 2005;280:14656. doi: 10.1074/jbc.M412585200. [DOI] [PubMed] [Google Scholar]
- 431.Dusaulcy R, Rancoule C, Gres S, Wanecq E, Colom A, Guigne C, van Meeteren LA, Moolenaar WH, Valet P, Saulnier-Blache JS. J Lipid Res. 2011;52:1247. doi: 10.1194/jlr.M014985. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 432.Gobeil F, Bernier SG, Vazquez-Tello A, Brault S, Beauchamp MH, Quiniou C, Marrache AM, Checchin D, Sennlaub F, Hou X, Nader M, Bkaily G, Ribeiro-da-Silva A, Goetzl EJ, Chemtob S. J Biol Chem. 2003;278:38875. doi: 10.1074/jbc.M212481200. [DOI] [PubMed] [Google Scholar]
- 433.Gillies L, Lee SC, Long JS, Ktistakis N, Pyne NJ, Pyne S. Cell Signal. 2009;21:675. doi: 10.1016/j.cellsig.2009.01.023. [DOI] [PubMed] [Google Scholar]
