Abstract
Sphingosine-1-phosphate (S1P) is a bioactive lipid mediator with crucial roles in a wide variety of cellular functions across a broad range of organisms. Though a simple molecule in structure, S1P functions are complex. The formation of S1P is catalyzed by one of two sphingosine kinases that have differential cellular distributions as well as both overlapping and opposing functions and which are activated by many different stimuli. S1P can act on a family of G protein-coupled receptors (S1PRs) that are also differentially expressed in different cell types, which influences the cellular responses to S1P. In addition to acting on receptors located on the plasma membrane, S1P can also function inside the cell, independently of S1PRs. It also appears that both the intracellular location and the isotype of sphingosine kinase involved are major determinants of inside-out signaling of S1P in response to many extracellular stimuli. This chapter is focused on the current literature on extracellular and intracellular actions of S1P
Keywords: sphingosine kinase, sphingosine-1-phosphate, inside-out signaling, nucleus, sphingosine, dihydrosphingosine, S1PR
1. Introduction
In just over a decade, the sphingolipid metabolite, sphingosine-1-phosphate (S1P), has emerged as a key regulator of numerous physiological functions, including cell growth and survival, angiogenesis, cell motility and migration, and lymphocyte trafficking 1. S1P promotes cell growth and inhibits apoptosis, while its precursors, ceramide and sphingosine, typically inhibit cell growth and induce apoptosis 1. Therefore, the cellular balance of these three sphingolipid metabolites, the “sphingolipid rheostat”, is of crucial importance in regulating cell fate 1. Moreover, sphingolipid metabolism has been found to be dysregulated in many human diseases, including cancer, inflammation, atherosclerosis, and asthma 2. S1P is produced intracellularly by two sphingosine kinases (SphK1 and SphK2), and is the ligand for a family of five G protein-coupled receptors, termed S1P1-5. However, there are some actions of S1P that appear to be independent of the known S1PRs. This review will focus on the emerging evidence for S1P as a second messenger.
2. Sphingolipid metabolism
Sphingolipids are ubiquitous components of the lipid bilayer of eukaryotic cells. Like glycerolipids, numerous agonists regulate sphingolipid metabolism to generate signaling molecules, including ceramide (N-acyl sphingosine), sphingosine and sphingosine-1-phosphate (S1P) 1, 3, 4. Ceramide, the backbone of all sphingolipids, is produced both by de novo synthesis and by turnover of sphingolipids, such as hydrolysis of sphingomyelin by sphingomyelinases. De novo synthesis at the endoplasmic reticulum (ER) is initiated by condensation of serine and palmitoyl-CoA catalyzed by serine palmitoyltransferase. The 3-ketosphinganine formed is rapidly reduced to dihydrosphingosine, which is subsequently N-acylated by one of a family of six (dihydro)ceramide synthases (CerS, formerly referred to as LASS) to form dihydroceramide, with the CerSs having differing but overlapping preferences for acyl chains from 16 to 26 carbons long. Finally, a 4–5 trans double bond is introduced in the sphingoid base to produce ceramide. Ceramide is then trafficked from the ER to the Golgi, where a variety of head groups are added, forming sphingolipids. After removal of these head groups during catabolism, deacylation of ceramide by ceramidases yields sphingosine, the most common sphingoid base in mammals. It is important to note that sphingosine and dihydrosphingosine (sphinganine) are not produced de novo and are only formed by catabolism of sphingolipids. These sphingoid bases can be re-utilized for complex sphingolipid biosynthesis or phosphorylated by two sphingosine kinases (SphK1 and SphK2) to form S1P. S1P can be degraded either by reversible dephosphorylation to sphingosine by phosphatases, including lysosomal phosphatases, members of the LPP family of lipid-specific phosphatases, and two S1P-specific phosphatases, termed SPP1 and SPP2 (reviewed in 5), or degraded by irreversible cleavage to ethanolamine phosphate and hexadecenal by S1P lyase (SPL) 6.
Ceramide and sphingosine, the precursors of S1P, are important regulatory components of stress responses, typically inducing growth arrest and apoptosis 3, 4. In contrast, S1P has been implicated in motility and cytoskeletal rearrangements, formation of adherens junctions, proliferation, survival, angiogenesis, and the trafficking of immune cells 1, 7–9. Thus, the dynamic balance between S1P and ceramide, and the consequent regulation of opposing signaling pathways, is an important factor that determines cell fate 10. S1P produces many of its effects by acting as a ligand for one or more of its five receptors, discussed below. However, although intracellular targets for both ceramide and sphingosine have been identified, indicating they are both bona fide second messengers, intracellular targets for S1P have remained elusive.
3. Sphingosine kinases
Oddly, like mammalian SphKs, two SphK isoenzymes are also found in organisms as diverse as yeast, slime molds, worms, flies, and mammals 7. In mammals, both SphKs have a broad and overlapping tissue distribution, with SphK1 predominating in lung and spleen, and SphK2 predominating in the heart, brain, and liver 11, 12. Both SphKs are members of the diacylglycerol kinase family, containing five conserved domains responsible for substrate binding and catalytic activity 13. Differential activation of SphKs can be determined by in vitro assays, as detergents stimulate SphK1 and inhibit SphK212. Conversely, SphK2 is stimulated by high salt, whereas SphK1 is inhibited. SphK1 and SphK2 have similar endogenous substrate specificities, with both being able to phosphorylate d-erythro-dihydrosphingosine and d-erythro-sphingosine, the two mammalian sphingoid bases 11, 13. Cells contain both S1P and dihydro-S1P, and both activate cell surface S1PRs with essentially the same affinity (see below). S1P predominates both in cells and plasma, and thus for the purposes of this review “S1P” will refer to all phosphorylated sphingoid bases, including phyto-S1P found in yeast and plants, unless otherwise indicated. SphK1 and SphK2 are primarily cytosolic, although their distributions are altered in different cell types and by various signals (see below). Homozygous single knockouts of either isoenzyme are viable and there are also no obvious phenotypes in yeast 14 or mice 15, 16, suggesting that they may have redundant, overlapping, or compensatory functions. Intriguingly, there is no functional redundancy in Drosophila, as deletion of Sk2 but not Sk1 results in flight defects and reduced fecundity 17. In yeast, even the double knockouts are viable 14. However, when Sphk1−/− and Sphk2−/− mice were crossed, mice lacking 1 to 3 of the Sphk alleles appeared normal, but mice lacking all four alleles died in utero due to defective brain and cardiovascular system development 16. This suggests that SphK1 and SphK2 are redundant in mammals, at least for viability. However, subtle yet significant phenotypic differences in Sphk1−/− and Sphk2−/− mice are emerging, and the apparent functional redundancy may not apply to a variety of pathophysiological conditions, suggesting that isozyme-specific targeting of SphKs may be an effective means of disease control or prevention.
3.1 SphK1
SphK1a is a 48 kDa splice variant that was cloned utilizing peptide sequences from SphK1 purified to homogeneity from rat kidneys 11. Most investigations of the role of SphK1 have focused on SphK1a, the major splice form, although two N-terminal extension splice variants have been described in humans, SphK1b (+14 aa) and SphK1c (+86 aa), all with similar enzymatic properties 18. SphK1 isoforms are predominantly cytosolic but have slightly different subcellular distributions, with SphK1b and SphK1c having greater plasma membrane localization 19. Interestingly, extracellular secretion of SphK1a, by unknown mechanisms through a Golgi-independent route has been described 19, although the biological significance of this is not yet clear. SphK1 was initially purified in part based on its ability to bind to a calcium-calmodulin column 20. Subsequently, it was shown that SphK1 has a calmodulin binding site between residues 191–206, the mutation of which blocks agonist-induced SphK1 translocation to the plasma membrane, but not its activation 21. An important advance in understanding regulation of SphK1 was the demonstration that SphK1 is phosphorylated on Ser225 by ERK1/222, which was necessary but not sufficient for its targeting to the plasma membrane 23. SphK1 has been shown to physically interact with a number of proteins that may affect its cellular localization, including RPK11824, PECAM-125, Acy126, and δ-catenin/NPRAP 27, among others. SphK1 is activated by diverse stimuli, including hormones, growth factors, immunoglobulin receptor crosslinking, cytokines, chemokines, and lysolipids, including S1P 1. Functionally, numerous studies have demonstrated a role for S1P produced by SphK1 in protection of cells from apoptosis, promotion of cell growth, stimulation of motility and tumorigenesis, and as an essential component of many signaling pathways, activating kinases, phospholipases, and inducing calcium release (reviewed in 1, 8, 28, 29). Indeed, many of the pro-growth and anti-apoptotic effects observed by exogenous addition of S1P can be reproduced by overexpression of SphK1.
3.2 SphK2
Much less is known about SphK2, which was cloned in mammalian cells based on its homology to SphK112. SphK2 has two splice variants. The smaller, originally cloned SphK2-S, contains 618 amino acids, and the N-terminally extended SphK2-L, consists of 654 amino acids 30. SphK2-L is expressed in human cells but not in mice, and is reported to be the predominant SphK2 mRNA in most cell lines and tissues except brain and kidney 30. SphK1 and SphK2 have similar abilities to phosphorylate endogenous substrates, but SphK2 is mainly responsible for phosphorylation of the immunosuppressive pro-drug, FTY72015, 31, 32. SphK2 was found to be a two hybrid interactor with the cytoplasmic domain of the IL-12 receptor β1, and SphK2 overexpression enhanced IL-12-mediated signaling 33. SphK2 also binds calmodulin at a site that is conserved between the SphKs, although unlike SphK1, interaction with calmodulin does not alter SphK2 localization 21. SphK2 contains a putative BH3 domain that has been shown to contribute to the ability of overexpressed SphK2 to induce apoptosis in a variety of cell types 34. In contrast to SphK1, only a few agonists have been reported to activate SphK2, including EGF 35, IgE receptor crosslinking 36, and IL-1β and TNFα37. Of note, EGF activates SphK2 and induces phosphorylation on Ser351 and Thr578, both dependent on ERK138. Moreover, this phosphorylation was required for SphK2-enhanced cell migration towards EGF 38. Intriguingly, although SphK2 expression typically inhibits growth, promotes apoptosis, and chemosensitizes several cell types 30, 34, 39, 40, SphK2 protects MCF-7 breast cancer cells and HCT116 colon cancer cells from doxorubicin-induced apoptosis by a pathway mediated by p53-independent upregulation of p2141.
3.3 SphK1 vs. SphK2
Even though mice with single knockouts of Sphk1 and Sphk2 are viable with few obvious phenotypes, suggesting that the SphKs have redundant functions, the kinases exhibit many differences in a variety of experimental systems. Indeed, several studies have shown that the two SphKs in yeast, Lcb3p and Lcb4p, do not complement one another (e.g. 42), although this may be due to the much higher activity of Lcb4p. Moreover, both SphKs, in yeast as well as mammals, have overlapping but distinct subcellular localizations, suggesting that they may interact with different proteins and/or lipids and utilize different sphingoid base substrate pools. The first clues that eukaryotic SphK1 and SphK2 may differ functionally came from the observations that, in contrast to many reports demonstrating a pro-growth, anti-apoptotic role for SphK1, SphK2 overexpression induced growth arrest and cell death 30, 34, 39. Consistent with these results, SphK1 decreased, while SphK2 increased, the sensitivity of several different cell lines to a variety of chemotherapeutic drugs 34, 40. However, other studies have revealed that SphK2 knockdown reduced proliferation of glioblastoma cell lines 43. The cell culture results demonstrating a role for SphK1 in cell growth and apoptosis are likely pathophysiologically relevant, as SphK1 is overexpressed in a number solid tumors, including breast, ovary, kidney, brain, stomach, and kidney 43, 44. Perhaps because of its role as a promoter of apoptosis, SphK2 but not SphK1 was reported to be responsible for the secretion of S1P during apoptosis of Jurkat T cells induced by staurosporin 45. Conversely, in hematopoietic cancer cells, it was recently shown that SphK1 produced the S1P that was secreted in response to doxorubicin-induced apoptosis 46.
The differential effects of the two SphKs on cell fate are due in part to their different roles in regulating ceramide levels. SphK1 expression decreases ceramide levels, likely by inhibiting ceramide synthases 47, 48. Conversely, SphK2 expression increases ceramide levels by increasing the salvage of sphingoid bases 47. In a pathway that is conserved in yeast 42, exogenous sphingoid bases must first be phosphorylated by a specific SphK (Lcb4p or SphK2), then dephosphorylated by a specific SPP (Lcb3p or SPP1) 49, 50. This cycle enables cells to re-acylate sphingoid bases to ceramides and likely represents a control point that cells use to regulate the amount of ceramide and sphingolipids at the levels of de novo synthesis (SphK1) and salvage of sphingoid bases (SphK2). SphK1 and SphK2 also appear to have different roles in the uptake of S1P, with SphK1 but not SphK2 cooperating in another phosphorylation/dephosphorylation cycle that utilizes cell surface LPP-1 to promote sphingoid base uptake51.
Differential effects of SphK1 and SphK2 are also apparent in transduction of signals from cell surface receptors. Perhaps because it was cloned first and thus is more well studied, SphK1-dependent formation of S1P has been found to be an important component of numerous receptor signaling pathways, but even in cases where participation of both SphK1 and SphK2 have been examined, it is still SphK1 that is typically implicated. To cite a few examples, estradiol activates SphK1, but not SphK2, ultimately leading to EGFR activation in MCF-7 cells 52; similarly, VEGF-induced activation of ERK1/2 in T24 bladder cancer cells requires SphK1 but not SphK253; moreover, targeting SphK1 but not SphK2 with siRNA also blocks TNFα-induced COX-2 induction and PGE2 secretion 54.
Many studies have demonstrated a role for S1P and S1PRs in cell motility (reviewed in 55), and SphK1, but not SphK2, has often been reported to be the source of S1P. For example, EGF stimulates the activity and translocation of SphK1 to lamellipodia in MCF-7 cells, and overexpression of SphK1 enhances migration towards EGF 56. EGF also has been shown to stimulate both SphK1 and SphK2 in HEK 293 cells, but only SphK1 was required for EGF-induced motility 35. The lack of requirement for SphK2 in these cells is the more remarkable because SphK2 is already localized at the plasma membrane, and suggests that a SphK may need to be targeted to a specific sub-compartment of the plasma membrane, perhaps in close proximity to its substrate and specific S1PRs and/or perhaps S1P transporter(s). Intriguingly, EGF also stimulates both SphK1 and SphK2 in MDA-MB-453 cells, but in these breast cancer cells, both SphKs are required for EGF-induced motility 35.
The roles of S1P in the immune system are complex and deciphering which SphK isozyme is involved is a challenging task. It has been shown that SphK1, but not SphK2, is required for degranulation of rat RBL-2H3 mast cells in response to IgE receptor crosslinking 57. In contrast, in mouse bone marrow-derived mast cells, both SphK1 and SphK2 are activated by IgE receptor crosslinking in a Fyn-dependent manner 36. Interestingly, SphK1 and SphK2 have different requirements for effectors downstream of Fyn, suggesting other levels of regulation of SphK activation. These authors also showed that exogenous S1P could only partially restore degranulation to Fyn−/− mice, hinting at an intracellular role for S1P (discussed below). This group later took advantage of Sphk1−/− and Sphk2−/− double knockout mice to demonstrate that in fetal liver-derived and bone marrow-derived mast cells, only SphK2 was responsible for IgE receptor triggered degranulation and cytokine release 58. However, in a passive systemic anaphylaxis model, they found that Sphk2−/− mice fared as well or slightly worse than wild-type mice in terms of increased plasma histamine levels. Conversely, Sphk1−/− mice had reduced plasma histamine levels. Sphk1−/− mice also had reduced plasma S1P levels compared to wild type and Sphk2−/− mice, and the intensity of the histamine released positively correlated with circulating S1P levels. The triple allele knockout Sphk1+/−Sphk2−/− mice had the lowest histamine responses and had plasma S1P levels as low as the Sphk1−/− mice. Thus, mast cell function in mice is determined both by SphK2 in mast cells (intrinsic S1P) and circulating S1P levels determined by non-mast cell SphK1 (extrinsic S1P). In contrast, in both human LAD2 mast cells and human umbilical cord blood-derived mast cells, knockdown of SphK1 expression decreased degranulation, cytokine release, and motility in response to IgE/antigen 59. Conversely, SphK2 was dispensable for antigen-induced degranulation, motility, or release of most cytokines 59. S1P likely also plays important roles in other types of immune cells, although neutrophil function in cells isolated from Sphk1−/− or Sphk2−/− mice, or even in the whole animals themselves, showed little observable differences between the knockouts and wild type. The Sphk2−/− mice did have increased disease progression in a lung infection model 60. Differences between mouse and human immune systems remain to be resolved, but will likely require SphK isozyme-specific inhibitors to elucidate the roles for the SphK1 and SphK2 in mast cell functions and development.
4. S1P Receptors
S1P is a ligand for five specific GPCRs, S1P1-5, formerly called endothelial differentiation gene (EDG) receptors, which are differentially expressed in different tissues. The cell type specific expression of S1PRs, as well as their differential coupling to different G proteins, explains the diverse signaling of S1P 61. As mentioned above, many stimuli, including hormones, immunoglobulin receptor ligation, growth factors, and cytokines, activate cytosolic SphKs and the production of S1P that is required for the full activity of these agonists. In many cases, the S1P produced activates cell surface S1PRs in a paracrine and/or autocrine manner (reviewed in 62). Indeed, many of the downstream effects of these stimuli require transactivation of one or more S1PRs, also called “inside-out” signaling. For example, in MCF-7 cells, estradiol stimulates ERK1/2 though a mechanism that requires at least two autocrine signaling loops 52. In the first loop, estradiol stimulates SphK1 and formation of S1P leading to activation of S1P3. In the second loop, S1P3 activates the metalloproteinase MMP-9, which in turn releases EGF from the EGF-heparin binding protein and activates EGFR, finally leading to ERK1/2 phosphorylation 52. How transactivation of S1PRs is accomplished is an intriguing puzzle as the SphKs and S1P production are both on the cytosolic side of the plasma membrane and S1PRs bind S1P on the exoplasmic side. While it has been suggested that SphK proteins themselves may be secreted and produce S1P extracellularly 19, it has been convincingly shown that the ABCC1 transporter mediates secretion of intracellularly produced S1P from mast cells 63. Moreover, S1PR transactivation is not only regulated by S1P secretion, cellular levels of S1P are also important, as decreasing levels by overexpression of SPP1 inhibits transactivation 64, while decreasing S1P degradation by inhibition of SPL promotes it 65. Such inside-out transactivation loops may be a general phenomenon, as chemotactic signals for neutrophils acting through their receptors induce secretion of ATP that then locally activates cell surface nucleotide receptors to coordinate directed cell migration 66.
4.1 S1P1
S1P1 is ubiquitously expressed, with high levels in brain, lung, spleen, cardiovascular system, and kidney. It was originally identified as an orphan GPCR involved in differentiation of endothelial cells 67. Since its discovery, many of the important physiological functions of S1P have been attributed to ligation of this receptor. It is now known that S1P1 plays a key role in angiogenesis, because its deletion in mice is embryonic lethal due to hemorrhage resulting from incomplete vascular maturation as smooth muscle cells and pericytes fail to migrate and envelop nascent endothelial tubes 68. Endothelial cell conditional S1P1 knockout mice have been generated using the Cre/Lox system, and these mice display the same vascular deficiencies, suggesting that S1P1 receptors on endothelial cells are also responsible for vessel coverage by smooth muscle cells 69. S1P1 also plays an important role in maintenance of endothelial and epithelial barrier integrity by functioning in conjunction with S1P2 and S1P3 to increase vascular integrity 70–72. Disruption of endothelial barriers leads to increased vascular permeability, often found in tumors and in inflammation. Silencing of S1P1 expression with siRNA blocks barrier enhancement, determined by transendothelial monolayer electrical resistance, while silencing of S1P3 inhibits vascular disruption 71. These experiments also identified the downstream signaling molecules Akt and Rac as effectors of S1P1 actions on vascular integrity 71. A role for S1P1 in vascular integrity has also been demonstrated in vivo 73–76, and experiments using a S1P1 selective antagonist demonstrated that S1P1 is crucial in maintaining vascular tone 77. Of particular interest are the conditional SphK1/SphK2 double knockout mice, which have normal vascular integrity despite having over 100-fold lower plasma S1P levels than wild type mice 78, suggesting that even a very low level of S1P is sufficient as long as S1P1 expression is normal.
S1P1 is also intimately involved in immune cell function. In particular, its expression is required for lymphocyte egress from lymph nodes. Resting T and B cells express primarily S1P1, and its downregulation or deletion results in lymphopenia due to the inability of lymphocytes to exit from the lymph nodes 15, 79. Additionally, transplantation of S1P1 deficient thymocytes and lymphocytes into normal mice results in their sequestration in lymph nodes and Peyer’s patches. Moreover, T-cells overexpressing S1P1 preferentially distribute into blood 80. These results confirm that S1P1 controls lymphocyte recirculation.
4.2 S1P2
S1P2 is also widely expressed in a variety of different cell types. Unlike S1P1 knockout mice, newborn S1P2 deficient mice do not demonstrate any striking abnormalities, although they have been reported to develop sporadic seizures between 3–7 weeks of age 81. Neocortical pyramidal cells from these mice also display an increase in excitability 81. In addition, S1P2−/− mice are deaf indicating that S1P2 is required for proper development of the auditory and vestibular systems 82, 83. S1P2 is also required for proper degranulation of mast cells 57. S1P2 is generally considered to be a repellant receptor as its activation inhibits cell migration and appears to work in opposition to S1P1 and S1P3, which both enhance cell migration 84. Similarly, activation of S1P2 activates ROCK/Rho and leads to increases in vascular permeability 72. Finally, although the visceral organs of S1P2−/− mice develop normally, expression of S1P2 promotes liver tissue remodeling in response to acute injury 85.
4.3 S1P3
Much like the lack of phenotypic effects of deletion of S1P2, deletion of S1P3 in mice does not generate any obvious phenotype. S1P3 is expressed in the cardiovascular system, lungs, kidney, intestines, spleen, and cartilage 86. Knockouts of both S1P2 and S1P3 increases perinatal lethality, but not to a great extent 87. However, the triple knockout of S1P1-3 leads to embryonic lethality due to massive vascular deficiencies perhaps even worse than those resulting from knockout of S1P1 alone 88. S1P3 is also an important regulator of vascular permeability signaling through the downstream effectors ROCK and Rho 72. A clear role for S1P3 has also been demonstrated in the regulation of heart rate 89, as S1P3 expression is localized to myocytes and perivascular smooth muscle cells, and its activation results in bradycardia and hypertension.
4.2 S1P4 and S1P5
S1P4-5 have much narrower patterns of expression than the dominant S1P1-3 receptors, localizing in human leukocytes, NK cells, airway smooth muscle cells and white matter of CNS tracts 90–93. S1P4 is primarily expressed in lymphoid tissues, including the thymus, spleen, bone marrow, appendix, and peripheral leukocytes 94. S1P4 directly couples to Gαi and Gα12/13 subunits of trimeric G proteins, and Jurkat cells overexpressing S1P4 display enhanced pertussis toxin-sensitive cell motility in the absence of S1P 95. S1P4 stimulation also activates the mitogen activated-protein kinases ERK1/2, activates phospholipase C, and modulates the opening of intracellular calcium stores 96, 97. Stimulation of S1P4 ectopically expressed on CHO-K1 cells induced cytoskeletal rearrangements and cell rounding, as well as its internalization following S1P stimulation 95. Whether or not S1P4 has a role in cell motility remains unclear. In D10G4.1 mouse Th2 cells and EL4.IL-2 mouse T cells lacking endogenous S1P receptors but transfected with S1P4, its activation failed to transduce chemotactic responses 98. These cells also displayed enhanced secretion of IL-10 and decreased proliferation in response to S1P 98.
S1P5 is highly expressed in oligodendrocytes 92; however, silencing of S1P5 expression does not inhibit myelination or produce any other obvious phenotype in these cells 99. Binding of S1P to S1P5 induces phosphatase-dependent inhibition of ERK1/2, resulting in an anti-proliferative phenotype 91, 100, 101. In addition, stimulation of rat oligodendrocytes with PDGF increases S1P1 expression with a concomitant downregulation of S1P5, resulting in an amplified mitogenic response 102. A recent report emonstrated that S1P5 is present in natural killer cells (NK). Mice deficient in S1P5 display aberrant NK cell homing and mobilization of NK cells to inflamed organs 93. Finally, despite a multitude of studies focusing on physiological functions of S1PRs, some actions of S1P resulting from activation of SphKs are independent of S1PRs.
5. Evidence for intracellular targets of S1P
As discussed above, cellular levels of S1P are controlled both by its synthesis and by its degradation. S1P can be degraded either by dephosphorylation back to sphingosine or irreversibly degraded by SPL to ethanolamine phosphate and fatty aldehyde. In fact, the latter is the only pathway in eukaryotic cells for degradation of sphingoid bases. Thus, S1P formation and subsequent degradation by SPL is one means for decreasing sphingolipid levels within the cell. Intriguingly, S1P is also an intermediate in the formation of sphingolipids from salvaged sphingoid bases. Though these pathways regulating sphingolipid levels demonstrate a central role for S1P, emerging evidence from yeast, plants, and mammals points to S1P as a classical, intracellular second messenger.
5.1 S1P in Saccharomyces cerevisiaei
The yeast genome does not encode a recognizable cell surface receptor for S1P and exogenous S1P does not affect yeast growth 103. Thus, any role for S1P in yeast physiology must therefore be intracellular. While no direct target for S1P has been found, S1P does indeed have intracellular functions in yeast. First, it has been demonstrated that yeast cells deleted of S1P phosphatase (lcb3/ysr2/lbp1) and SPL (dpl1) accumulate large amounts of S1P and are non-viable or very slow growing. This growth arrest can be can be rescued if the major yeast SphK (lcb4) is deleted, indicating that intracellular S1P suppresses yeast cell growth 104, 105. Indeed, sphingosine itself induces growth arrest in yeast, and this effect can be blocked by mutational inactivation of SphK 103. The growth inhibition may be due to elevated levels cytosolic calcium, as S1P has been shown to increase intracellular calcium levels 106. In contrast, while heat shock-induced cell cycle arrest is mediated by the sphingoid base itself 107, Heat shock also increases SphK activity 103 and S1P accumulation 108, suggesting a functional role for S1P in heat shock responses. Preventing S1P metabolism by deletion of either the S1P phosphatase Lcb3p 109, 110 or SPL 108 led to enhanced levels of S1P and increased heat shock tolerance. Moreover, mutational inactivation of SphK reversed the protective effect of SPL deletion 103. That deletion of either an S1P phosphatase or SPL promotes heat tolerance indicates that S1P has direct actions, rather than merely functioning as a metabolic intermediate. Identification of molecular targets of S1P in yeast would aid this quest in mammalian cells.
5.2 S1P in Arabidopsis thaliana
Abscisic acid is a plant hormone responsible for mediating responses to drought conditions such as closure of stomata to prevent water loss. It was recently demonstrated that drought increased S1P levels in plants and that abscisic acid-induced stomatal closure was reduced by SphK inhibitors 111. Subsequently, it was shown that abscisic acid activated SphK in Arabidopsis 112. Intriguingly, these authors showed that the effects of S1P on stomatal openings were dependent on the single canonical Gα protein in plants, GPA1. This finding suggests that S1P might be acting through plant GPCRs. However, Arabidopsis has only one GPCR-like protein, designated GCR1, which is not homologous to the known S1PRs, does not bind phosphorylated sphingoid bases, and GCR1 mutants are hypersensitive to S1P-induced stomatal closure 113. Thus, S1P likely regulates stomatal apertures and drought responses intracellularly in plants.
5.3 S1P in mammalian cells
The observation that expression of SphK1, but not SphK2, decreases ceramide levels and increases dihydrosphingosine levels 47 suggests that S1P produced by SphK1 may negatively regulate one or more of the six (dihydro)ceramide synthases (CerS), leading to accumulation of its substrate, dihydrosphingosine. Subsequently, using lysates from cells over-expressing individual CerSs, S1P was shown to be a non-competitive in vitro inhibitor of CerS2, but not of other CerSs 48. In silico analysis identified two domains in CerS2 with predicted homology to S1PRs, and mutation of one arginine to alanine in each of these two domains removed S1P inhibition without altering CerS activity 48. The authors concluded that S1P directly binds to, and inhibits, CerS2. However, it is also possible that S1P acts through intermediate proteins in cell lysates to inhibit CerS2. Moreover, CerS2 does not use C16 or C18 acyl CoAs as substrates, yet it is C16 and C18 ceramides that are most affected by expression of SphK147. Additionally, S1P produced by SphK1 reverses CerS1-induced chemosensitivity, suggesting that S1P directly affects CerS240. Finally, it is not yet clear whether inhibition of CerS2 by S1P is a signal-mediated effect or whether it is simply feedback product inhibition. Further work is needed to confirm that S1P binds to and inhibits CerS2.
As mentioned previously, autocrine and/or paracrine transactivation of S1PRs is typically accompanied by translocation of SphK1 to the plasma membrane. However, both SphK1 and SphK2 have been shown to translocate to other cellular compartments, suggesting that S1P may be produced locally in these compartments to act on specific intracellular targets. For example, SphK1 is targeted to internal membranes through interaction with RPK11824 and Acy126. SphK1 translocated to nascent phagosomes promotes maturation into mature phagolysosomes 114. SphK1 has also been shown to translocate to the nuclear envelope during S-phase 115, and this translocation may play a role in the ability of SphK1 overexpression to promote the G1/S transition 116. Conversely, SphK2 has been shown to reside in the nucleoplasm in certain cells, where it functions to arrest cells 30, 39. Moreover, SphK2 has a nuclear export signal sequence that is activated by in G1 phorbol ester-induced phosphorylation, likely through protein kinase D 117. Whether the nuclear export signal serves to promote SphK2/S1P signaling in the cytosol or to decrease SphK2/S1P signaling in the nucleus is unclear. In some cells, SphK2 has been shown to translocate to the ER under stress conditions and promote apoptosis 47. Indeed, targeting SphK1 to the ER induced apoptosis 34, suggesting that S1P produced at the ER has specific targets.
A direct target for S1P in the ER has not conclusively been identified, although strong evidence indicates that S1P can activate thapsigargin-sensitive calcium channels, likely in the ER. S1P induced inositol trisphosphate receptor-independent release of calcium from permeablized cells 118 and from cell fractions rich in rough, but not smooth, ER 119. Consistent with these results, cells overexpressing SphK2, which localizes in part to the ER, were also shown to have elevated intracellular calcium 47. Conversely, fetal liver derived-mast cells from Sphk2−/− mice have a defect in calcium mobilization in response to IgE receptor crosslinking that cannot be restored with exogenous S1P 58, again suggesting an internal S1P target. Exogenous S1P increases calcium in HEK293 cells, a response that can be inhibited with pertussis toxin. However, microinjection of S1P increases calcium bypassing a pertussis toxin block, again supporting a role for intracellular S1P in calcium release 120. Similarly, caged S1P can elicit calcium mobilization in cells that do not respond to exogenous S1P 121. Likewise, UTP stimulates calcium mobilization in a SphK-dependent manner, but this effect is not mimicked by exogenous S1P 122. Finally, exogenous S1P itself can stimulate SphK, and SphK inhibitors reduced calcium release induced by S1P 123. Intracellular calcium release by S1P may be an evolutionarily ancient pathway, as it has been shown that S1P can increase calcium in yeast 106. In sum, these results strongly suggest that S1P is a second messenger that can activate calcium channels.
Finally, there are other effects of S1P that cannot be explained by activation of the known S1PRs. For example, overexpression of SphK1 promotes survival of endothelial cells in part through PECAM-1 expression and activation of Akt, an effect that is not reproduced by exogenous S1P 124. Dihydro-S1P and S1P bind and activate S1PRs with similar affinities, but dihydro-S1P cannot recapitulate all of the effects of S1P 125–128. For example, S1P, but not dihydro-S1P, protects male germ cells from apoptosis, an effect that was linked to inhibition of NF-κB and activation of Akt 128. A similar protection pattern was observed for HL-60 and PC-12 cells 125. Conversely, S1P-phosphonate, which does not bind to S1PRs, also protects these cells from apoptosis 125, suggesting an intracellular action. Other studies using embryonic fibroblasts from S1PR knockout mice ruled out S1PR involvement, demonstrating that SphK1 overexpression stimulated growth and survival in wild type and in S1PR negative cells 129. In the APCmin/+ model of intestinal tumorigenesis, it was shown that Sphk1−/− mice, but not S1p1−/+, S1p2−/−, or S1p3−/− mice, had reduced tumor progression and size 130, suggesting an intracellular role for S1P. Moreover, SphK1 null mice had elevated levels of sphingosine but not S1P, so it is possible that reduced tumor progression and size was due to inhibitory effects of sphingosine rather than to the absence of S1P stimulation. S1PR knockouts were similarly used to demonstrate that SphK2-induced apoptosis was S1PR independent 34. Consistent with a role for SphK2 in apoptosis, the FTY720 analog AAL(R), which is phosphorylated by SphK2 but only poorly by SphK1, induces apoptosis in Jurkat cells and primary splenocytes 131. The authors showed that AAL(R) had to be phosphorylated to affect cells as the non-phosphorylated AAL(R) isomer did not induce apoptosis in cells from Sphk2−/− mice. However, exogenous addition of phosphorylated AAL(R) did not induce apoptosis, suggesting both that phospho-AAL(R) must be generated at or near its site of action and that S1PRs are not involved in SphK2-induced apoptosis.
Intracellular S1P has also been linked to regulation of inflammatory responses. CD4+ T cells from SphK2 knockout mice displayed a hyperactivated phenotype, increased proliferation, and enhanced secretion of cytokines and STAT5 activation in response to IL-2132. This phenotype was physiologically relevant, as T cells from SphK2 knockout mice induced a much more rapid response than T cells from wild type littermates in an adoptive transfer model of inflammatory bowel disease. The hyperresponsiveness to IL-2 could not be reversed with exogenous S1P, indicating that internal S1P normally suppresses IL-2-induced inflammatory responses. Interestingly, activation of the T cell receptor in Th1 and Th2 cells leads to increased expression of SphK1, but not SphK2, and SphK1 negatively regulates chemokine expression, although the authors did not examine whether exogenous S1P reproduced chemokine suppression 133. Similarly, it has been shown that in primary umbilical vein endothelial cells, the pro-inflammatory cytokine TNFα greatly increased expression of the S1P phosphatase SPP2134. Induction of SPP2 was required for the TNFα-induced production of IL-1β and IL-8. SPP2 is an integral membrane protein of the ER and its requirement for induction of inflammatory responses suggests that it acts at the ER to remove an inhibitory S1P signal. Moreover, downregulation of SPP1 demonstrated that it played no role in TNFα-induced cytokine secretion. As SPP1 is also an ER resident protein, these results suggest a role for S1P at a specific subcompartment of the ER. Moreover, parallel findings have been reported in yeast, where the S1P-specific phosphatases, YSR2 and YSR3, are both localized to the ER and have overlapping but distinct functions 5.
Many studies have utilized non-isozyme specific SphK inhibitors to implicate S1P in various signaling pathways (e.g. 135, 136), leading to the conclusion that S1P was acting intracellularly in these systems, but S1P release was not detected. Still, care should be used in interpreting these results as S1P release may have been below the limits of detection. Moreover, exogenous S1P can activate all surface S1PRs, perhaps resulting in net opposing effects. It is also possible that signaling events localize secretion of S1P to regions of the plasma membrane where specific receptors to be activated are localized. Thus, lack of detectable S1P secretion does not definitively show that S1PRs are not involved in a particular signaling pathway.
Furthermore, studies in endothelial cells using a pan sphingosine kinase inhibitor demonstrated that endogenously generated S1P functions as a positive modulator of calcium entry via store operated channels (SOC), whereas exogenously administered S1P initiated calcium release from the ER similar to histamine and decreased endothelial cell permeability 137. These results suggest that the production of intracellular S1P, and not the secretion of S1P to act on other immunoregulatory cells, is the primary determinant of the inflammatory response.
Recently, a new study demonstrated that the vascular permeability inhibitor Ang-1 stimulated SphK1 and led to increases in intracellular S1P and decreases in vascular permeability. This function remained undisturbed even when expression of S1P1-3 were downregulated 138. Exogenous S1P also decreased vascular permeability, but not in cells where S1PRs were downregulated 138. These results suggest that Ang-1/SphK1 actions on vascular permeability are mediated via an intracellular mechanism.
6. Implications and future directions
It is now clear that the bioactive lipid mediator S1P exerts effects both intracellularly and extracellularly. In addition, SphK1 and SphK2 are not only distributed differently throughout tissues and within cells, S1P produced by these kinases can have different, sometimes opposite, downstream effects. How can one simple molecule like S1P have such a wide range of effects? It seems that in the case of S1P, the location of its production may be a major determinant of the resulting phenotype. The key to unlocking this riddle will be the inevitable discovery of intracellular S1P binding partners, but to date, none have been unequivocally identified. Extensive studies of proteins in the nucleus, particularly those involved in the transcriptional machinery and the regulation of the cell cycle, are clearly indicated to elucidate the mechanism by which S1P exerts its effects there. Additionally, the consequences of activation or inhibition SphK1 vs. SphK2 must be studied with more scrutiny, as it is apparent that despite catalyzing the same reaction, they have different functions. In addition, because the intracellular actions of S1P involve many functions related to cancer and other diseases, a full understanding of both the extracellular and intracellular actions of S1P will be needed to best design clinical therapies targeting SphKs and S1P production as well as its receptors.
Acknowledgments
Supported by National Institutes of Health Grants R37GM043880, RO1CA61774, RO1AI050094 and U19AI077435-018690 (SS), NIH National Research Service Award F30NS058008 (GS), and the NIMH Intramural Research Program (SM).
References
- 1.Spiegel S, Milstien S. Sphingosine-1-phosphate: an enigmatic signalling lipid. Nature Rev Mol Cell Boil. 2003;4:397–407. doi: 10.1038/nrm1103. [DOI] [PubMed] [Google Scholar]
- 2.Tamable K, Pugh SW, Milstien S, et al. “Inside-out” signaling of sphingosine-1-phosphate: therapeutic targets. Pharmacology Rev. 2008;60:181–195. doi: 10.1124/pr.107.07113. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Ogre men B, Han nun YA. Biologically active sphingolipids in cancer pathogenesis and treatment. Nature Rev Cancer. 2004;4:604–616. doi: 10.1038/nrc1411. [DOI] [PubMed] [Google Scholar]
- 4.Reynolds CP, Maurer BJ, Lovesick RN. Ceramide synthesis and metabolism as a target for cancer therapy. Cancer Let. 2004;206:169–180. doi: 10.1016/j.canlet.2003.08.034. [DOI] [PubMed] [Google Scholar]
- 5.Maceyka M, Milstien S, Spiegel S. Measurement of mammalian sphingosine-1-phosphate phosphohydrolase activity in vitro and in vivo. Methods Enzymol. 2007;434:243–256. doi: 10.1016/S0076-6879(07)34013-5. [DOI] [PubMed] [Google Scholar]
- 6.Bandhuvula P, Saba JD. Sphingosine-1-phosphate lyase in immunity and cancer: silencing the siren. Trends Mol Med. 2007;13:210–217. doi: 10.1016/j.molmed.2007.03.005. [DOI] [PubMed] [Google Scholar]
- 7.Maceyka M, Payne SG, Milstien S, et al. Sphingosine kinase, sphingosine-1-phosphate, and apoptosis. Biochim Biophys Acta. 2002;1585:193–201. doi: 10.1016/s1388-1981(02)00341-4. [DOI] [PubMed] [Google Scholar]
- 8.Saba JD, Hla T. Point-counterpoint of sphingosine 1-phosphate metabolism. Circ Res. 2004;94:724–734. doi: 10.1161/01.RES.0000122383.60368.24. [DOI] [PubMed] [Google Scholar]
- 9.Cyster JG. Chemokines, sphingosine-1-phosphate, and cell migration in secondary lymphoid organs. Annu Rev Immunol. 2005;23:127–159. doi: 10.1146/annurev.immunol.23.021704.115628. [DOI] [PubMed] [Google Scholar]
- 10.Cuvillier O, Pirianov G, Kleuser B, et al. Suppression of ceramide-mediated programmed cell death by sphingosine-1-phosphate. Nature. 1996;381:800–803. doi: 10.1038/381800a0. [DOI] [PubMed] [Google Scholar]
- 11.Kohama T, Olivera A, Edsall L, et al. Molecular cloning and functional characterization of murine sphingosine kinase. J Biol Chem. 1998;273:23722–23728. doi: 10.1074/jbc.273.37.23722. [DOI] [PubMed] [Google Scholar]
- 12.Liu H, Sugiura M, Nava VE, et al. Molecular cloning and functional characterization of a novel mammalian sphingosine kinase type 2 isoform. J Biol Chem. 2000;275:19513–19520. doi: 10.1074/jbc.M002759200. [DOI] [PubMed] [Google Scholar]
- 13.Liu H, Chakravarty D, Maceyka M, et al. Sphingosine kinases: a novel family of lipid kinases. Prog Nucl Acid Res. 2002;71:493–511. doi: 10.1016/s0079-6603(02)71049-0. [DOI] [PubMed] [Google Scholar]
- 14.Nagiec MM, Skrzypek M, Nagiec EE, et al. The LCB4 (YOR171c) and LCB5 (YLR260w) genes of Saccharomyces encode long chain base kinases. J Biol Chem. 1998;273:19437–19442. doi: 10.1074/jbc.273.31.19437. [DOI] [PubMed] [Google Scholar]
- 15.Allende ML, Sasaki T, Kawai H, et al. Mice deficient in sphingosine kinase 1 are rendered lymphopenic by FTY720. J Biol Chem. 2004;279:52487–52492. doi: 10.1074/jbc.M406512200. [DOI] [PubMed] [Google Scholar]
- 16.Mizugishi K, Yamashita T, Olivera A, et al. Essential role for sphingosine kinases in neural and vascular development. Mol Cell Biol. 2005;25:11113–11121. doi: 10.1128/MCB.25.24.11113-11121.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Herr DR, Fyrst H, Creason MB, et al. Characterization of the Drosophila sphingosine kinases and requirement for Sk2 in normal reproductive function. J Biol Chem. 2004;279:12685–12694. doi: 10.1074/jbc.M310647200. [DOI] [PubMed] [Google Scholar]
- 18.Billich A, Bornancin F, Devay P, et al. Phosphorylation of the imunomodulatory drug FTY720 by sphingosine kinases. J Biol Chem. 2003;278:47408–47415. doi: 10.1074/jbc.M307687200. [DOI] [PubMed] [Google Scholar]
- 19.Venkataraman K, Thangada S, Michaud J, et al. Extracellular export of sphingosine kinase-1a contributes to the vascular S1P gradient. Biochem J. 2006;397:461–471. doi: 10.1042/BJ20060251. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Olivera A, Kohama T, Tu Z, et al. Purification and characterization of rat kidney sphingosine kinase. J Biol Chem. 1998;273:12576–12583. doi: 10.1074/jbc.273.20.12576. [DOI] [PubMed] [Google Scholar]
- 21.Sutherland CM, Moretti PA, Hewitt NM, et al. The calmodulin-binding site of sphingosine kinase and its role in agonist-dependent translocation of sphingosine kinase 1 to the plasma membrane. J Biol Chem. 2006;281:11693–11701. doi: 10.1074/jbc.M601042200. [DOI] [PubMed] [Google Scholar]
- 22.Pitson SM, Moretti PA, Zebol JR, et al. Activation of sphingosine kinase 1 by ERK1/2-mediated phosphorylation. EMBO J. 2003;22:5491–5500. doi: 10.1093/emboj/cdg540. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Pitson SM, Xia P, Leclercq TM, et al. Phosphorylation-dependent translocation of sphingosine kinase to the plasma membrane drives its oncogenic signalling. J Exp Med. 2005;201:49–54. doi: 10.1084/jem.20040559. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Hayashi S, Okada T, Igarashi N, et al. Identification and characterization of RPK118, a novel sphingosine kinase-1-binding protein. J Biol Chem. 2002;277:33319–33324. doi: 10.1074/jbc.M201442200. [DOI] [PubMed] [Google Scholar]
- 25.Fukuda Y, Aoyama Y, Wada A, et al. Identification of PECAM-1 association with sphingosine kinase 1 and its regulation by agonist-induced phosphorylation. Biochim Biophys Acta. 2004;1636:12–21. doi: 10.1016/j.bbalip.2003.11.006. [DOI] [PubMed] [Google Scholar]
- 26.Maceyka M, Nava VE, Milstien S, et al. Aminoacylase 1 is a sphingosine kinase 1-interacting protein. FEBS Lett. 2004;568:30–34. doi: 10.1016/j.febslet.2004.04.093. [DOI] [PubMed] [Google Scholar]
- 27.Fujita T, Okada T, Hayashi S, et al. Delta-catenin/NPRAP (neural plakophilin-related armadillo repeat protein) interacts with and activates sphingosine kinase 1. Biochem J. 2004;382:717–723. doi: 10.1042/BJ20040141. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Hait NC, Oskeritzian CA, Paugh SW, et al. Sphingosine kinases, sphingosine 1-phosphate, apoptosis and diseases. Biochim Biophys Acta. 2006;1758:2016–2026. doi: 10.1016/j.bbamem.2006.08.007. [DOI] [PubMed] [Google Scholar]
- 29.Hannun YA, Obeid LM. Principles of bioactive lipid signalling: lessons fromsphingolipids. Nat Rev Mol Cell Biol. 2008;9:139–150. doi: 10.1038/nrm2329. [DOI] [PubMed] [Google Scholar]
- 30.Okada T, Ding G, Sonoda H, et al. Involvement of N-terminal-extended form of sphingosine kinase 2 in serum-dependent regulation of cell proliferation and apoptosis. J Biol Chem. 2005;280:36318–36325. doi: 10.1074/jbc.M504507200. [DOI] [PubMed] [Google Scholar]
- 31.Paugh SW, Payne SG, Barbour SE, et al. The immunosuppressant FTY720 is phosphorylated by sphingosine kinase type 2. FEBS Lett. 2003;554:189–193. doi: 10.1016/s0014-5793(03)01168-2. [DOI] [PubMed] [Google Scholar]
- 32.Sanchez T, Estrada-Hernandez T, Paik JH, et al. Phosphorylation and action of the immunomodulator FTY720 inhibits VEGF-induced vascular permeability. J Biol Chem. 2003;278:47281–47290. doi: 10.1074/jbc.M306896200. [DOI] [PubMed] [Google Scholar]
- 33.Yoshimoto T, Furuhata M, Kamiya S, et al. Positive modulation of IL-12 signaling by sphingosine kinase 2 associating with the IL-12 receptor beta1 cytoplasmic region. J Immunol. 2003;171:1352–1359. doi: 10.4049/jimmunol.171.3.1352. [DOI] [PubMed] [Google Scholar]
- 34.Liu H, Toman RE, Goparaju S, et al. Sphingosine kinase type 2 is a putative BH3-only protein that induces apoptosis. J Biol Chem. 2003;278:40330–40336. doi: 10.1074/jbc.M304455200. [DOI] [PubMed] [Google Scholar]
- 35.Hait NC, Sarkar S, Le Stunff H, et al. Role of sphingosine kinase 2 in cell migration towards epidermal growth factor. J Biol Chem. 2005;280:29462–29469. doi: 10.1074/jbc.M502922200. [DOI] [PubMed] [Google Scholar]
- 36.Olivera A, Urtz N, Mizugishi K, et al. IgE-dependent activation of sphingosine kinases 1 and 2 and secretion of sphingosine 1-phosphate requires Fyn kinase and contributes to mast cell responses. J Biol Chem. 2006;281:2515–2525. doi: 10.1074/jbc.M508931200. [DOI] [PubMed] [Google Scholar]
- 37.Mastrandrea LD, Sessanna SM, Laychock SG. Sphingosine kinase activity and sphingosine-1 phosphate production in rat pancreatic islets and INS-1 cells: response to cytokines. Diabetes. 2005;54:1429–1436. doi: 10.2337/diabetes.54.5.1429. [DOI] [PubMed] [Google Scholar]
- 38.Hait NC, Bellamy A, Milstien S, et al. Sphingosine kinase type 2 activation by ERK-mediated phosphorylation. J Biol Chem. 2007;282:12058–12065. doi: 10.1074/jbc.M609559200. [DOI] [PubMed] [Google Scholar]
- 39.Igarashi N, Okada T, Hayashi S, et al. Sphingosine kinase 2 is a nuclear protein and inhibits DNA synthesis. J Biol Chem. 2003;278:46832–46839. doi: 10.1074/jbc.M306577200. [DOI] [PubMed] [Google Scholar]
- 40.Min J, Mesika A, Sivaguru M, et al. (Dihydro)ceramide synthase 1 regulated sensitivity to cisplatin is associated with the activation of p38 mitogen-activated protein kinase and is abrogated by sphingosine kinase 1. Mol Cancer Res. 2007;5:801–812. doi: 10.1158/1541-7786.MCR-07-0100. [DOI] [PubMed] [Google Scholar]
- 41.Sankala HM, Hait NC, Paugh SW, et al. Involvement of sphingosine kinase 2 in p53-independent induction of p21 by the chemotherapeutic drug doxorubicin. Cancer Res. 2007;67:10466–10474. doi: 10.1158/0008-5472.CAN-07-2090. [DOI] [PubMed] [Google Scholar]
- 42.Funato K, Lombardi R, Vallee B, et al. Lcb4p is a key regulator of ceramide synthesis from exogenous long chain sphingoid base in Saccharomyces cerevisiae. J Biol Chem. 2003;278:7325–7334. doi: 10.1074/jbc.M209925200. [DOI] [PubMed] [Google Scholar]
- 43.Van Brocklyn JR, Jackson CA, Pearl DK, et al. Sphingosine kinase-1 expression correlates with poor survival of patients with glioblastoma multiforme: roles of sphingosine kinase isoforms in growth of glioblastoma cell lines. J Neuropathol Exp Neurol. 2005;64:695–705. doi: 10.1097/01.jnen.0000175329.59092.2c. [DOI] [PubMed] [Google Scholar]
- 44.French KJ, Schrecengost RS, Lee BD, et al. Discovery and evaluation of inhibitors of human sphingosine kinase. Cancer Res. 2003;63:5962–5969. [PubMed] [Google Scholar]
- 45.Weigert A, Johann AM, von Knethen A, et al. Apoptotic cells promote macrophage survival by releasing the antiapoptotic mediator sphingosine-1-phosphate. Blood. 2006;108:1635–1642. doi: 10.1182/blood-2006-04-014852. [DOI] [PubMed] [Google Scholar]
- 46.Gude DR, Alvarez SE, Paugh SW, et al. Apoptosis induces expression of sphingosine kinase 1 to release sphingosine-1-phosphate as a “come-and-get-me” signal. FASEB J. 2008;22:2629–2638. doi: 10.1096/fj.08-107169. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Maceyka M, Sankala H, Hait NC, et al. Sphk1 and Sphk2: Sphingosine kinase isoenzymes with opposing functions in sphingolipid metabolism. J Biol Chem. 2005;280:37118–37129. doi: 10.1074/jbc.M502207200. [DOI] [PubMed] [Google Scholar]
- 48.Laviad EL, Albee L, Pankova-Kholmyansky I, et al. Characterization of ceramide synthase 2: tissue distribution, substrate specificity, and inhibition by sphingosine 1-phosphate. J Biol Chem. 2008;283:5677–5684. doi: 10.1074/jbc.M707386200. [DOI] [PubMed] [Google Scholar]
- 49.Le Stunff H, Galve-Roperh I, Peterson C, et al. Sphingosine-1-phosphate phosphohydrolase in regulation of sphingolipid metabolism and apoptosis. J Cell Biol. 2002;158:1039–1049. doi: 10.1083/jcb.200203123. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Le Stunff H, Giussani P, Maceyka M, et al. Recycling of sphingosine is regulated by the concerted actions of sphingosine-1-phosphate phosphohydrolase 1 and sphingosine kinase 2. J Biol Chem. 2007;282:3472–3480. doi: 10.1074/jbc.M703329200. [DOI] [PubMed] [Google Scholar]
- 51.Zhao Y, Kalari SK, Usatyuk PV, et al. Intracellular generation of sphingosine 1-phosphate in human lung endothelial cells: Role of lipid phosphate phosphatase-1 and sphingosine kinase 1. J Biol Chem. 2007;282:14165–14177. doi: 10.1074/jbc.M701279200. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Sukocheva O, Wadham C, Holmes A, et al. Estrogen transactivates EGFR via the sphingosine 1-phosphate receptor Edg-3: the role of sphingosine kinase-1. J Cell Biol. 2006;173:301–310. doi: 10.1083/jcb.200506033. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Shu X, Wu W, Mosteller RD, et al. Sphingosine kinase mediates vascular endothelial growth factor-induced activation of ras and mitogen-activated protein kinases. Mol Cell Biol. 2002;22:7758–7768. doi: 10.1128/MCB.22.22.7758-7768.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Pettus BJ, Bielawski J, Porcelli AM, et al. The sphingosine kinase 1/sphingosine-1-phosphate pathway mediates COX-2 induction and PGE2 production in response to TNF-alpha. FASEB J. 2003;17:1411–1421. doi: 10.1096/fj.02-1038com. [DOI] [PubMed] [Google Scholar]
- 55.Donati C, Bruni P. Sphingosine 1-phosphate regulates cytoskeleton dynamics: Implications in its biological response. Biochim Biophys Acta. 2006;1758:2037–2048. doi: 10.1016/j.bbamem.2006.06.015. [DOI] [PubMed] [Google Scholar]
- 56.Sarkar S, Maceyka M, Hait NC, et al. Sphingosine kinase 1 is required for migration, proliferation and survival of MCF-7 human breast cancer cells. FEBS Lett. 2005;579:5313–5317. doi: 10.1016/j.febslet.2005.08.055. [DOI] [PubMed] [Google Scholar]
- 57.Jolly PS, Bektas M, Olivera A, et al. Transactivation of sphingosine-1-phosphate receptors by Fc{epsilon}RI triggering is required for normal mast cell degranulation and chemotaxis. J Exp Med. 2004;199:959–970. doi: 10.1084/jem.20030680. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Olivera A, Mizugishi K, Tikhonova A, et al. The sphingosine kinase-sphingosine-1-posphate axis is a determinant of mast cell function and anaphylaxis. Immunity. 2007;26:287–297. doi: 10.1016/j.immuni.2007.02.008. [DOI] [PubMed] [Google Scholar]
- 59.Oskeritzian CA, Alvarez SE, Hait NC, et al. Distinct roles of sphingosine kinases 1 and 2 in human mast-cell functions. Blood. 2008;111:4193–4200. doi: 10.1182/blood-2007-09-115451. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Zemann B, Urtz N, Reuschel R, et al. Normal neutrophil functions in sphingosine kinase type 1 and 2 knockout mice. Immunol Lett. 2007;109:56–63. doi: 10.1016/j.imlet.2007.01.001. [DOI] [PubMed] [Google Scholar]
- 61.Brinkmann V. Sphingosine 1-phosphate receptors in health and disease: mechanistic insights from gene deletion studies and reverse pharmacology. Pharmacol Ther. 2007;115:84–105. doi: 10.1016/j.pharmthera.2007.04.006. [DOI] [PubMed] [Google Scholar]
- 62.Alvarez SE, Milstien S, Spiegel S. Autocrine and paracrine roles of sphingosine-1-phosphate. Trends Endocrinol Metab. 2007;18:300–307. doi: 10.1016/j.tem.2007.07.005. [DOI] [PubMed] [Google Scholar]
- 63.Mitra P, Oskeritzian CA, Payne SG, et al. Role of ABCC1 in export of sphingosine-1-phosphate from mast cells. Proc Natl Acad Sci USA. 2006;103:16394–16399. doi: 10.1073/pnas.0603734103. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Le Stunff H, Mikami A, Giussani P, et al. Role of sphingosine-1-phosphate phosphatase 1 in epidermal growth factor-induced chemotaxis. J Biol Chem. 2004;279:34290–34297. doi: 10.1074/jbc.M404907200. [DOI] [PubMed] [Google Scholar]
- 65.Schwab SR, Pereira JP, Matloubian M, et al. Lymphocyte sequestration through S1P lyase inhibition and disruption of S1P gradients. Science. 2005;309:1735–1739. doi: 10.1126/science.1113640. [DOI] [PubMed] [Google Scholar]
- 66.Chen Y, Corriden R, Inoue Y, et al. ATP release guides neutrophil chemotaxis via P2Y2 and A3 receptors. Science. 2006;314:1792–1795. doi: 10.1126/science.1132559. [DOI] [PubMed] [Google Scholar]
- 67.Hla T, Maciag T. An abundant transcript induced in differentiating human endothelial cells encodes a polypeptide with structural similarities to G-protein coupled receptors. J Biol Chem. 1990;265:9308–9313. [PubMed] [Google Scholar]
- 68.Liu Y, Wada R, Yamashita T, et al. Edg-1, the G protein-coupled receptor for sphingosine-1-phosphate, is essential for vascular maturation. J Clin Invest. 2000;106:951–961. doi: 10.1172/JCI10905. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Allende ML, Yamashita T, Proia RL. G-protein coupled receptor S1P1 acts within endothelial cells to regulate vascular maturation. Blood. 2003;102:3665–3667. doi: 10.1182/blood-2003-02-0460. [DOI] [PubMed] [Google Scholar]
- 70.McVerry BJ, Garcia JG. Endothelial cell barrier regulation by sphingosine 1-phosphate. J Cell Biochem. 2004;92:1075–1085. doi: 10.1002/jcb.20088. [DOI] [PubMed] [Google Scholar]
- 71.Singleton PA, Dudek SM, Ma SF, et al. Transactivation of sphingosine 1-phosphate receptors is essential for vascular barrier regulation. Novel role for hyaluronan and CD44 receptor family. J Biol Chem. 2006;281:34381–34393. doi: 10.1074/jbc.M603680200. [DOI] [PubMed] [Google Scholar]
- 72.Sanchez T, Skoura A, Wu MT, et al. Induction of vascular permeability by the sphingosine-1-phosphate receptor-2 (S1P2R) and its downstream effectors ROCK and PTEN. Arterioscler Thromb Vasc Biol. 2007;27:1312–1318. doi: 10.1161/ATVBAHA.107.143735. [DOI] [PubMed] [Google Scholar]
- 73.Singleton PA, Dudek SM, Chiang ET, et al. Regulation of sphingosine 1-phosphate-induced endothelial cytoskeletal rearrangement and barrier enhancement by S1P1 receptor, PI3 kinase, Tiam1/Rac1, and alpha-actinin. FASEB J. 2005;19:1646–1656. doi: 10.1096/fj.05-3928com. [DOI] [PubMed] [Google Scholar]
- 74.Finigan JH, Dudek SM, Singleton PA, et al. Activated protein C mediates novel lung endothelial barrier enhancement: Role of sphingosine 1-phosphate receptor transactivation. J Biol Chem. 2005;280:17286–17293. doi: 10.1074/jbc.M412427200. [DOI] [PubMed] [Google Scholar]
- 75.McVerry BJ, Garcia JG. In vitro and in vivo modulation of vascular barrier integrity by sphingosine 1-phosphate: mechanistic insights. Cell Signal. 2005;17:131–139. doi: 10.1016/j.cellsig.2004.08.006. [DOI] [PubMed] [Google Scholar]
- 76.Lee JF, Zeng Q, Ozaki H, et al. Dual roles of tight junction associated protein, zonula occludens-1, in sphingosine-1-phosphate mediated endothelial chemotaxis and barrier integrity. J Biol Chem. 2006;281:29190–29200. doi: 10.1074/jbc.M604310200. [DOI] [PubMed] [Google Scholar]
- 77.Sanna MG, Wang SK, Gonzalez-Cabrera PJ, et al. Enhancement of capillary leakage and restoration of lymphocyte egress by a chiral S1P(1) antagonist in vivo. Nat Chem Biol. 2006;2:434–441. doi: 10.1038/nchembio804. [DOI] [PubMed] [Google Scholar]
- 78.Pappu R, Schwab SR, Cornelissen I, et al. Promotion of lymphocyte egress into blood and lymph by distinct sources of sphingosine-1-phosphate. Science. 2007;316:295–298. doi: 10.1126/science.1139221. [DOI] [PubMed] [Google Scholar]
- 79.Matloubian M, Lo CG, Cinamon G, et al. Lymphocyte egress from thymus and peripheral lymphoid organs is dependent on S1P receptor 1. Nature. 2004;427:355–360. doi: 10.1038/nature02284. [DOI] [PubMed] [Google Scholar]
- 80.Chi H, Flavell RA. Cutting edge: regulation of T cell trafficking and primary immune responses by sphingosine 1-phosphate receptor 1. J Immunol. 2005;174:2485–2488. doi: 10.4049/jimmunol.174.5.2485. [DOI] [PubMed] [Google Scholar]
- 81.MacLennan AJ, Carney PR, Zhu WJ, et al. An essential role for the H218/AGR16/Edg-5/LP(B2) sphingosine 1-phosphate receptor in neuronal excitability. Eur J Neurosci. 2001;14:203–209. doi: 10.1046/j.0953-816x.2001.01634.x. [DOI] [PubMed] [Google Scholar]
- 82.Kono M, Belyantseva IA, Skoura A, et al. Deafness and stria vascularis defects in S1P2 receptor null mice. J Biol Chem. 2007;282:10690–10696. doi: 10.1074/jbc.M700370200. [DOI] [PubMed] [Google Scholar]
- 83.Herr DR, Chun J. Effects of LPA and S1P on the nervous system and implications for their involvement in disease. Curr Drug Targets. 2007;8:155–167. doi: 10.2174/138945007779315669. [DOI] [PubMed] [Google Scholar]
- 84.Lepley D, Paik JH, Hla T, et al. The G protein-coupled receptor S1P2 regulates Rho/Rho kinase pathway to inhibit tumor cell migration. Cancer Res. 2005;65:3788–3795. doi: 10.1158/0008-5472.CAN-04-2311. [DOI] [PubMed] [Google Scholar]
- 85.Serriere-Lanneau V, Teixeira-Clerc F, Li L, et al. The sphingosine 1-phosphate receptor S1P2 triggers hepatic wound healing. FASEB J. 2007;21:2005–2013. doi: 10.1096/fj.06-6889com. [DOI] [PubMed] [Google Scholar]
- 86.Ishii I, Friedman B, Ye X, et al. Selective loss of sphingosine 1-phosphate signaling with no obvious phenotypic abnormality in mice lacking its G protein-coupled receptor, LP(B3)/EDG-3. J Biol Chem. 2001;276:33697–33704. doi: 10.1074/jbc.M104441200. [DOI] [PubMed] [Google Scholar]
- 87.Ishii I, Ye X, Friedman B, et al. Marked perinatal lethality and cellular signaling deficits in mice Null for the two sphingosine 1-phosphate receptors, S1P2/LPB2/EDG-5 and S1P3/LPB3/EDG-3. J Biol Chem. 2002;277:25152–25159. doi: 10.1074/jbc.M200137200. [DOI] [PubMed] [Google Scholar]
- 88.Kono M, Mi Y, Liu Y, et al. The sphingosine-1-phosphate receptors S1P1, S1P2, and S1P3 function coordinately during embryonic angiogenesis. J Biol Chem. 2004;279:29367–29373. doi: 10.1074/jbc.M403937200. [DOI] [PubMed] [Google Scholar]
- 89.Forrest M, Sun SY, Hajdu R, et al. Immune cell regulation and cardiovascular efects of sphingosine 1-phosphate receptoragonists in rodents are mediated via distinct receptor subtypes. J Pharmacol Exp Ther. 2004;309:758–768. doi: 10.1124/jpet.103.062828. [DOI] [PubMed] [Google Scholar]
- 90.Graler MH, Bernhardt G, Lipp M. EDG6, a novel G-protein-coupled receptor related to receptors for bioactive lysophospholipids, is specifically expressed in lymphoid tissue. Genomics. 1998;53:164–169. doi: 10.1006/geno.1998.5491. [DOI] [PubMed] [Google Scholar]
- 91.Im DS, Heise CE, Ancellin N, et al. Characterization of a novel sphingosine 1-phosphate receptor, Edg-8. J Biol Chem. 2000;275:14281–14286. doi: 10.1074/jbc.275.19.14281. [DOI] [PubMed] [Google Scholar]
- 92.Terai K, Soga T, Takahashi M, et al. Edg-8 receptors are preferentially expressed in oligodendrocyte lineage cells of the rat CNS. Neuroscience. 2003;116:1053–1062. doi: 10.1016/s0306-4522(02)00791-1. [DOI] [PubMed] [Google Scholar]
- 93.Walzer T, Chiossone L, Chaix J, et al. Natural killer cell trafficking in vivo requires a dedicated sphingosine 1-phosphate receptor. Nat Immunol. 2007;8:1337–1344. doi: 10.1038/ni1523. [DOI] [PubMed] [Google Scholar]
- 94.Kluk MJ, Hla T. Signaling of sphingosine-1-phosphate via the S1P/EDG-family of G- protein-coupled receptors. Biochim Biophys Acta. 2002;1582:72–80. doi: 10.1016/s1388-1981(02)00139-7. [DOI] [PubMed] [Google Scholar]
- 95.Graler MH, Grosse R, Kusch A, et al. The sphingosine 1-phosphate receptor S1P4 regulates cell shape and motility via coupling to Gi and G12/13. J Cell Biochem. 2003;89:507–519. doi: 10.1002/jcb.10537. [DOI] [PubMed] [Google Scholar]
- 96.Van Brocklyn JR, Graler MH, Bernhardt G, et al. Sphingosine-1-phosphate is a ligand for the G protein-coupled receptor EDG-6. Blood. 2000;95:2624–2629. [PubMed] [Google Scholar]
- 97.Yamazaki Y, Kon J, Sato K, et al. Edg-6 as a putative sphingosine 1-phosphate receptor coupling to Ca(2+) signaling pathway. Biochem Biophys Res Commun. 2000;268:583–589. doi: 10.1006/bbrc.2000.2162. [DOI] [PubMed] [Google Scholar]
- 98.Wang W, Graeler MH, Goetzl EJ. Type 4 sphingosine 1-phosphate G protein-coupled receptor (S1P4) transduces S1P effects on T cell proliferation and cytokine secretion without signaling migration. FASEB J. 2005;19:1731–1733. doi: 10.1096/fj.05-3730fje. [DOI] [PubMed] [Google Scholar]
- 99.Jaillard C, Harrison S, Stankoff B, et al. Edg8/S1P5: an oligodendroglial receptor with dual function on process retraction and cell survival. J Neurosci. 2005;25:1459–1469. doi: 10.1523/JNEUROSCI.4645-04.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Gonda K, Okamoto H, Takuwa N, et al. The novel sphingosine 1-phosphate receptor AGR16 is coupled via pertussis toxin-sensitive and -insensitive G-proteins to multiple signalling pathways. Biochem J. 1999;337:67–75. [PMC free article] [PubMed] [Google Scholar]
- 101.Malek RL, Toman RE, Edsall LC, et al. Nrg-1 belongs to the endothelial differentiation gene family of G protein-coupled sphingosine-1-phosphate receptors. J Biol Chem. 2001;276:5692–5699. doi: 10.1074/jbc.M003964200. [DOI] [PubMed] [Google Scholar]
- 102.Jung CG, Kim HJ, Miron VE, et al. Functional consequences of S1P receptor modulation in rat oligodendroglial lineage cells. Glia. 2007;55:1656–1667. doi: 10.1002/glia.20576. [DOI] [PubMed] [Google Scholar]
- 103.Lanterman MM, Saba JD. Characterization of sphingosine kinase (SK) activity in Saccharomyces cerevisiae and isolation of SK-deficient mutants. Biochem J. 1998;332:525–531. doi: 10.1042/bj3320525. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Kim S, Fyrst H, Saba J. Accumulation of phosphorylated sphingoid long chain bases results in cell growth inhibition in Saccharomyces cerevisiae. Genetics. 2000;156:1519–1529. doi: 10.1093/genetics/156.4.1519. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.Zhang X, Skrzypek MS, Lester RL, et al. Elevation of endogenous sphingolipid long-chain base phosphates kills Saccharomyces cerevisiae cells. Curr Genet. 2001;40:221–233. doi: 10.1007/s00294-001-0259-6. [DOI] [PubMed] [Google Scholar]
- 106.Birchwood CJ, Saba JD, Dickson RC, et al. Calcium influx and signaling in yeast stimulated by intracellular sphingosine 1-phosphate accumulation. J Biol Chem. 2001;276:11712–11718. doi: 10.1074/jbc.M010221200. [DOI] [PubMed] [Google Scholar]
- 107.Jenkins GM, Hannun YA. Role for de novo sphingoid base biosynthesis in the heat-induced transient cell cycle arrest of Saccharomyces cerevisiae. J Biol Chem. 2001;276:8574–8581. doi: 10.1074/jbc.M007425200. [DOI] [PubMed] [Google Scholar]
- 108.Skrzypek MS, Nagiec MM, Lester RL, et al. Analysis of phosphorylated sphingolipid long-chain bases reveals potential roles in heat stress and growth control in Saccharomyces. J Bacteriol. 1999;181:1134–1140. doi: 10.1128/jb.181.4.1134-1140.1999. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.Mao C, Saba JD, Obeid LM. The dihydrosphingosine-1-phosphate phosphatases of Saccharomyces cerevisiae are important regulators of cell proliferation and heat stress responses. Biochem J. 1999;342:667–675. [PMC free article] [PubMed] [Google Scholar]
- 110.Mandala SM, Thornton R, Galve-Roperh I, et al. Molecular cloning and characterization of a lipid phosphohydrolase that degrades sphingosine-1-phosphate and induces cell death. Proc Natl Acad Sci USA. 2000;97:7859–7864. doi: 10.1073/pnas.120146897. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111.Ng CK, Carr K, McAinsh MR, et al. Drought-induced guard cell signal transduction involves sphingosine-1-phosphate. Nature. 2001;410:596–599. doi: 10.1038/35069092. [DOI] [PubMed] [Google Scholar]
- 112.Coursol S, Fan LM, Le Stunff H, et al. Sphingolipid signalling in Arabidopsis guard cells involves heterotrimeric G proteins. Nature. 2003;423:651–654. doi: 10.1038/nature01643. [DOI] [PubMed] [Google Scholar]
- 113.Pandey S, Assmann SM. The Arabidopsis putative G protein-coupled receptor GCR1 interacts with the G protein alpha subunit GPA1 and regulates abscisic acid signaling. Plant Cell. 2004;16:1616–1632. doi: 10.1105/tpc.020321. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114.Thompson CR, Iyer SS, Melrose N, et al. Sphingosine kinase 1 (SK1) is recruited to nascent phagosomes in human macrophages: inhibition of SK1 translocation by Mycobacterium tuberculosis. J Immunol. 2005;174:3551–3561. doi: 10.4049/jimmunol.174.6.3551. [DOI] [PubMed] [Google Scholar]
- 115.Kleuser B, Maceyka M, Milstien S, et al. Stimulation of nuclear sphingosine kinase activity by platelet-derived growth factor. FEBS Lett. 2001;503:85–90. doi: 10.1016/s0014-5793(01)02697-7. [DOI] [PubMed] [Google Scholar]
- 116.Olivera A, Kohama T, Edsall LC, et al. Sphingosine kinase expression increases intracellular sphingosine-1-phosphate and promotes cell growth and survival. J Cell Biol. 1999;147:545–558. doi: 10.1083/jcb.147.3.545. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117.Ding G, Sonoda H, Yu H, et al. Protein kinase D-mediated phosphorylation and nuclear export of sphingosine kinase 2. J Biol Chem. 2007;282:27493–27502. doi: 10.1074/jbc.M701641200. [DOI] [PubMed] [Google Scholar]
- 118.Mattie M, Brooker G, Spiegel S. Sphingosine-1-phosphate, a putative second messenger, mobilizes calcium from internal stores via an inositol trisphosphate-independent pathway. J Biol Chem. 1994;269:3181–3188. [PubMed] [Google Scholar]
- 119.Ghosh TK, Bian J, Gill DL. Sphingosine 1-phosphate generated in the endoplasmic reticulum membrane activates release of stored calcium. J Biol Chem. 1994;269:22628–22635. [PubMed] [Google Scholar]
- 120.Meyer zu Heringdorf D, Lass H, Alemany R, et al. Sphingosine kinase-mediated Ca2+ signalling by G-protein-coupled receptors. EMBO J. 1998;17:2830–2837. doi: 10.1093/emboj/17.10.2830. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121.Meyer zu Heringdorf D, Liliom K, Schaefer M, et al. Photolysis of intracellular caged sphingosine-1-phosphate causes Ca2+ mobilization independently of G-protein-coupled receptors. FEBS Lett. 2003;554:443–449. doi: 10.1016/s0014-5793(03)01219-5. [DOI] [PubMed] [Google Scholar]
- 122.Alemany R, Sichelschmidt B, zu Heringdorf DM, et al. Stimulation of sphingosine-1-phosphate formation by the P2Y(2) receptor in HL-60 cells: Ca(2+) requirement and implication in receptor-mediated Ca(2+) mobilization, but not MAP kinase activation. Mol Pharmacol. 2000;58:491–497. doi: 10.1124/mol.58.3.491. [DOI] [PubMed] [Google Scholar]
- 123.Blom T, Slotte JP, Pitson SM, et al. Enhancement of intracellular sphingosine-1-phosphate production by inositol 1,4,5-trisphosphate-evoked calcium mobilisation in HEK-293 cells: endogenous sphingosine-1-phosphate as a modulator of the calcium response. Cell Signal. 2005;17:827–836. doi: 10.1016/j.cellsig.2004.11.022. [DOI] [PubMed] [Google Scholar]
- 124.Limaye VS, Li X, Hahn C, et al. Sphingosine kinase-1 enhances endothelial cell survival through a PECAM-1-dependent activation of PI-3K/Akt and regulation of Bcl-2 family members. Blood. 2005;105:3169–3177. doi: 10.1182/blood-2004-02-0452. [DOI] [PubMed] [Google Scholar]
- 125.Van Brocklyn JR, Lee MJ, Menzeleev R, et al. Dual actions of sphingosine-1-phosphate: extracellular through the Gi-coupled orphan receptor edg-1 and intracellular to regulate proliferation and survival. J Cell Biol. 1998;142:229–240. doi: 10.1083/jcb.142.1.229. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126.Xia P, Gamble JR, Rye KA, et al. Tumor necrosis factor-a induces adhesion molecule expression through the sphingosine kinase pathway. Proc Natl Acad Sci USA. 1998;95:14196–14201. doi: 10.1073/pnas.95.24.14196. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127.Morita Y, Perez GI, Paris F, et al. Oocyte apoptosis is suppressed by disruption of the acid sphingomyelinase gene or by sphingosine-1-phosphate therapy. Nature Med. 2000;6:1109–1114. doi: 10.1038/80442. [DOI] [PubMed] [Google Scholar]
- 128.Suomalainen L, Pentikainen V, Dunkel L. Sphingosine-1-phosphate inhibits nuclear factor kappaB activation and germ cell apoptosis in the human testis independently of its receptors. Am J Pathol. 2005;166:773–781. doi: 10.1016/s0002-9440(10)62298-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129.Olivera A, Rosenfeldt HM, Bektas M, et al. Sphingosine kinase type 1 Induces G12/13-mediated stress fiber formation yet promotes growth and survival independent of G protein coupled receptors. J Biol Chem. 2003;278:46452–46460. doi: 10.1074/jbc.M308749200. [DOI] [PubMed] [Google Scholar]
- 130.Kohno M, Momoi M, Oo ML, et al. Intracellular role for sphingosine kinase 1 in intestinal adenoma cell proliferation. Mol Cell Biol. 2006;26:7211–7223. doi: 10.1128/MCB.02341-05. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131.Don AS, Martinez-Lamenca C, Webb WR, et al. Essential requirement for sphingosine kinase 2 in a sphingolipid apoptosis pathway activated by FTY720 analogs. J Biol Chem. 2007;282:15833–15842. doi: 10.1074/jbc.M609124200. [DOI] [PubMed] [Google Scholar]
- 132.Samy ET, Meyer CA, Caplazi P, et al. Cutting Edge: Modulation of Intestinal Autoimmunity and IL-2 Signaling by Sphingosine Kinase 2 Independent of Sphingosine 1-Phosphate. J Immunol. 2007;179:5644–5648. doi: 10.4049/jimmunol.179.9.5644. [DOI] [PubMed] [Google Scholar]
- 133.Yang J, Castle BE, Hanidu A, et al. Sphingosine kinase 1 Is a negative regulator of CD4+ Th1 cells. J Immunol. 2005;175:6580–6588. doi: 10.4049/jimmunol.175.10.6580. [DOI] [PubMed] [Google Scholar]
- 134.Mechtcheriakova D, Wlachos A, Sobanov J, et al. Sphingosine 1-phosphate phosphatase 2 is induced during inflammatory responses. Cell Signal. 2007;19:748–760. doi: 10.1016/j.cellsig.2006.09.004. [DOI] [PubMed] [Google Scholar]
- 135.Wang F, Van Brocklyn JR, Edsall L, et al. Sphingosine-1-phosphate inhibits motility of human breast cancer cells independently of cell surface receptors. Cancer Res. 1999;59:6185–6191. [PubMed] [Google Scholar]
- 136.Hammad SM, Crellin HG, Wu BX, et al. Dual and distinct roles for sphingosine kinase 1 and sphingosine 1 phosphate in the response to inflammatory stimuli in RAW macrophages. Prostaglandins Other Lipid Mediat. 2007;85:107–114. doi: 10.1016/j.prostaglandins.2007.11.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 137.Itagaki K, Yun JK, Hengst JA, et al. Sphingosine 1-phosphate has dual functions in the regulation of endothelial cell permeability and Ca2+ metabolism. J Pharmacol Exp Ther. 2007;323:186–191. doi: 10.1124/jpet.107.121210. [DOI] [PubMed] [Google Scholar]
- 138.Li X, Stankovic M, Bonder CS, et al. Basal and angiopoietin-1-mediated endothelial permeability is regulated by sphingosine kinase-1. Blood. 2008;111:3489–3497. doi: 10.1182/blood-2007-05-092148. [DOI] [PMC free article] [PubMed] [Google Scholar]