Home >> Research Area >>GPCR/G protein>>Sphingosine>> Sphingosine-1-phosphate

Sphingosine-1-phosphate

endogenous second messenger and ligand for S1PR1 CAS# 26993-30-6

Sphingosine-1-phosphate

Catalog No. BCC7034----Order now to get a substantial discount!

Product Name & Size Price Stock
Sphingosine-1-phosphate:1mg $179.00 In stock
Sphingosine-1-phosphate:2mg $304.00 In stock
Sphingosine-1-phosphate:5mg $716.00 In stock
Sphingosine-1-phosphate:10mg $1253.00 In stock
Related Products
  • JNK-IN-7

    Catalog No.:BCC1672
    CAS No.:1408064-71-0
  • CC-401 hydrochloride

    Catalog No.:BCC1458
    CAS No.:1438391-30-0
  • CEP 1347

    Catalog No.:BCC7982
    CAS No.:156177-65-0
  • AEG 3482

    Catalog No.:BCC8088
    CAS No.:63735-71-7
  • AS 602801

    Catalog No.:BCC1369
    CAS No.:848344-36-5
  • CC-930

    Catalog No.:BCC1459
    CAS No.:899805-25-5

Quality Control of Sphingosine-1-phosphate

Number of papers citing our products

Chemical structure

Sphingosine-1-phosphate

3D structure

Chemical Properties of Sphingosine-1-phosphate

Cas No. 26993-30-6 SDF Download SDF
PubChem ID 5283560 Appearance Powder
Formula C18H38NO5P M.Wt 379.48
Type of Compound N/A Storage Desiccate at -20°C
Solubility Soluble in Chloroform,Dichloromethane,Ethyl Acetate,DMSO,Acetone,etc.
Chemical Name [(E,2S,3R)-2-amino-3-hydroxyoctadec-4-enyl] dihydrogen phosphate
SMILES CCCCCCCCCCCCCC=CC(C(COP(=O)(O)O)N)O
Standard InChIKey DUYSYHSSBDVJSM-KRWOKUGFSA-N
Standard InChI InChI=1S/C18H38NO5P/c1-2-3-4-5-6-7-8-9-10-11-12-13-14-15-18(20)17(19)16-24-25(21,22)23/h14-15,17-18,20H,2-13,16,19H2,1H3,(H2,21,22,23)/b15-14+/t17-,18+/m0/s1
General tips For obtaining a higher solubility , please warm the tube at 37 ℃ and shake it in the ultrasonic bath for a while.Stock solution can be stored below -20℃ for several months.
We recommend that you prepare and use the solution on the same day. However, if the test schedule requires, the stock solutions can be prepared in advance, and the stock solution must be sealed and stored below -20℃. In general, the stock solution can be kept for several months.
Before use, we recommend that you leave the vial at room temperature for at least an hour before opening it.
About Packaging 1. The packaging of the product may be reversed during transportation, cause the high purity compounds to adhere to the neck or cap of the vial.Take the vail out of its packaging and shake gently until the compounds fall to the bottom of the vial.
2. For liquid products, please centrifuge at 500xg to gather the liquid to the bottom of the vial.
3. Try to avoid loss or contamination during the experiment.
Shipping Condition Packaging according to customer requirements(5mg, 10mg, 20mg and more). Ship via FedEx, DHL, UPS, EMS or other couriers with RT, or blue ice upon request.

Biological Activity of Sphingosine-1-phosphate

DescriptionEndogenous second messenger involved in the control of cell proliferation and motility, and Ca2+ mobilization. Acts as an agonist at sphingosine-1-phosphate receptors (S1P1-5) and as an activator of GPR3, GPR6 and GPR12. Effectors regulated include p38 MAP kinase, PLC, adenylyl cyclase, myosin light chain phosphatase and focal adhesion kinase.

Sphingosine-1-phosphate Dilution Calculator

Concentration (start)
x
Volume (start)
=
Concentration (final)
x
Volume (final)
 
 
 
C1
V1
C2
V2

calculate

Sphingosine-1-phosphate Molarity Calculator

Mass
=
Concentration
x
Volume
x
MW*
 
 
 
g/mol

calculate

Preparing Stock Solutions of Sphingosine-1-phosphate

1 mg 5 mg 10 mg 20 mg 25 mg
1 mM 2.6352 mL 13.1759 mL 26.3518 mL 52.7037 mL 65.8796 mL
5 mM 0.527 mL 2.6352 mL 5.2704 mL 10.5407 mL 13.1759 mL
10 mM 0.2635 mL 1.3176 mL 2.6352 mL 5.2704 mL 6.588 mL
50 mM 0.0527 mL 0.2635 mL 0.527 mL 1.0541 mL 1.3176 mL
100 mM 0.0264 mL 0.1318 mL 0.2635 mL 0.527 mL 0.6588 mL
* Note: If you are in the process of experiment, it's necessary to make the dilution ratios of the samples. The dilution data above is only for reference. Normally, it's can get a better solubility within lower of Concentrations.

Organizitions Citing Our Products recently

 
 
 

Calcutta University

University of Minnesota

University of Maryland School of Medicine

University of Illinois at Chicago

The Ohio State University

University of Zurich

Harvard University

Colorado State University

Auburn University

Yale University

Worcester Polytechnic Institute

Washington State University

Stanford University

University of Leipzig

Universidade da Beira Interior

The Institute of Cancer Research

Heidelberg University

University of Amsterdam

University of Auckland
TsingHua University
TsingHua University
The University of Michigan
The University of Michigan
Miami University
Miami University
DRURY University
DRURY University
Jilin University
Jilin University
Fudan University
Fudan University
Wuhan University
Wuhan University
Sun Yat-sen University
Sun Yat-sen University
Universite de Paris
Universite de Paris
Deemed University
Deemed University
Auckland University
Auckland University
The University of Tokyo
The University of Tokyo
Korea University
Korea University

Background on Sphingosine-1-phosphate

Sphingosine-1-phosphate is an endogenous second messenger that involved in cell proliferation and survival and is a ligand for S1PR1 [1] [2].

Sphingosine-1-phosphate receptor 1 (S1PR1), also known as endothelial differentiation gene 1 (EDG1), is a G-protein-coupled receptor for sphingosine-1-phosphate (S1P) and is involved in regulating endothelial cell cytoskeletal structure, capillary-like network formation, migration and vascular maturation.

Sphingosine-1-phosphate is an endogenous second messenger and a ligand for S1PR1. In endothelial cells, S1P (1-20 µM) induced EDG-l-dependent morphogenesis. In HEK293EDG-1 cells, S1P bound to S1PR1 with Kd value of 8.1 nM in a time-dependent way. In cells transfected with EDG-l, S1P activated ERK-1 and ERK-2 via EDG-1. S1P existed in platelets and released through platelet activation [2]. In human embryonic kidney (HEK) cells, S1P significantly increased [Ca2+]i with EC50 value of 2 nM in a pertussis toxin-sensitive way and inhibited cAMP accumulation induced by forskolin. Also, S1P increased the binding of guanosine 5’-3-O-(thio)triphosphate to cell membranes. In guinea pig atrial myocytes, S1P at the extracellular face activated Gi protein-regulated inwardly rectifying potassium channels [3]. S1P inhibited programmed cell death mediated by ceramide [4].

References:
[1].  Olivera A, Spiegel S. Sphingosine-1-phosphate as second messenger in cell proliferation induced by PDGF and FCS mitogens. Nature, 1993, 365(6446): 557-560.
[2].  Lee MJ, Van Brocklyn JR, Thangada S, et al. Sphingosine-1-phosphate as a ligand for the G protein-coupled receptor EDG-1. Science, 1998, 279(5356): 1552-1555.
[3].  van Koppen C, Meyer zu Heringdorf M, Laser KT, et al. Activation of a high affinity Gi protein-coupled plasma membrane receptor by sphingosine-1-phosphate. J Biol Chem, 1996, 271(4): 2082-2087.
[4].  Cuvillier O, Pirianov G, Kleuser B, et al. Suppression of ceramide-mediated programmed cell death by sphingosine-1-phosphate. Nature, 1996, 381(6585): 800-803.

Featured Products
New Products
 

References on Sphingosine-1-phosphate

Data on subgroup specific baseline characteristics and serum sphingosine-1-phosphate concentrations in the Study of Health in Pomerania.[Pubmed:28377994]

Data Brief. 2017 Mar 11;12:46-50.

In this data article, we provide subgroup specific baseline characteristics and serum Sphingosine-1-phosphate (S1P) concentrations for healthy individuals within the Study of Health in Pomerania (SHIP)-TREND cohort. After exclusion of subjects with cardiovascular disease, diabetes mellitus, hypertension, metabolic syndrome, elevated liver enzymes and/or chronic kidney disease stadium III or IV, four subgroups were defined according to different limits for body mass index (BMI), alterations in blood lipid levels and smoking status. Tables show respective clinical and laboratory parameters stratified by gender. Serum S1P concentrations are also stratified by age groups. The data presented herein is related to the research article entitled "Reference intervals for serum Sphingosine-1-phosphate in the population-based Study of Health in Pomerania" (E. Moritz, D. Wegner, S. Gross, M. Bahls, M. Dorr, S.B. Felix, T. Ittermann, S. Oswald, M. Nauck, N. Friedrich, R.H. Boger, G. Daum, E. Schwedhelm, B.H. Rauch, Clin Chim Acta. 468 (2017) 25-31) [1].

Demyelination induced by oxidative stress is regulated by sphingosine 1-phosphate receptors.[Pubmed:28375547]

Glia. 2017 Jul;65(7):1119-1136.

Oxidative stress is a pathological condition defined as an imbalance between production and removal of reactive oxygen species. This process causes structural cell damage, disrupts DNA repair and induces mitochondrial dysfunction. Many in vitro studies have used direct bolus application of H2 O2 to investigate the role of oxidative stress in cell culture. In this study, using mouse organotypic cerebellar slice cultures, the effects of H2 O2 -induced oxidative stress on myelination state were examined, using bolus concentrations of H2 O2 (0.1-1 mM) and low-continuous H2 O2 ( approximately 20 muM) generated from glucose oxidase and catalase (GOX-CAT). Using these models, the potential therapeutic effects of pFTY720, an oral therapy used in multiple sclerosis, was also examined. We found bolus treatment of H2 O2 (0.5 mM) and, for the first time, low-continuous H2 O2 (GOX-CAT) to induce demyelination in organotypic slices. Both bolus H2 O2 and GOX-CAT treatments significantly decreased vimentin expression in these slice cultures as well as increased cell death in isolated astrocyte cultures. Importantly, pre-treatment with pFTY720 significantly attenuated both bolus H2 O2 and GOX-CAT-induced demyelination and the GOX-CAT-induced decrease in vimentin in cerebellar slices, without altering levels of the proinflammatory cytokines such as IL-6 and CX3CL1. We also observed increased SMI-32 immunoreactivity in the white matter tract induced by GOX-CAT indicating axonal damage, which was remarkably attenuated by pFTY720. Taken together, this data establishes a novel GOX-CAT model of demyelination and demonstrates that pFTY720 can act independently of inflammatory cytokines to attenuate decreases in vimentin, as well as axonal damage and demyelination induced by oxidative stress.

Activation mechanisms of the first sphingosine-1-phosphate receptor.[Pubmed:28370663]

Protein Sci. 2017 Jun;26(6):1150-1160.

Activation of the first Sphingosine-1-phosphate receptor (S1PR1 ) promotes permeability of the blood brain barrier, astrocyte and neuronal protection, and lymphocyte egress from secondary lymphoid tissues. Although an agonist often activates the S1PR1 , the receptor exhibits high levels of basal activity. In this study, we performed long-timescale molecular dynamics and accelerated molecular dynamics (aMD) simulations to investigate activation mechanisms of the ligand-free (apo) S1PR1 . In the aMD enhanced sampling simulations, we observed four independent events of activation, which is characterized by close interaction between Y311(7.53) and Y221(5.58) and increased distance between the intracellular ends of transmembrane (TM) helices 3 and 6. Although TM helices TM3, TM6, TM5 and, TM7 are associated with GPCR activation, we discovered that their movements are not necessarily correlated during activation. Instead, TM5 showed a decreased correlation with each of these regions during activation. During activation of the apo receptor, Y221(5.58) and Y311(7.53) became more solvated, because a water channel formed in the intracellular pocket. Additionally, a lipid molecule repeatedly entered the receptor between the extracellular ends of TM1 and TM7, providing important insights into the pathway of ligand entry into the S1PR1 .

In vivo intrabursal administration of bioactive lipid sphingosine-1-phosphate enhances vascular integrity in a rat model of ovarian hyperstimulation syndrome.[Pubmed:28379469]

Mol Hum Reprod. 2017 Jun 1;23(6):417-427.

STUDY QUESTION: Can the bioactive lipid sphingosine-1 phosphate (S1P) act as an endothelial barrier-enhancing molecule and, in turn, restore the vascular integrity and homoeostasis in a rat model of ovarian hyperstimulation syndrome (OHSS). STUDY ANSWER: In vivo administration of S1P may prevent the early onset of OHSS and decrease its severity. WHAT IS KNOWN ALREADY: Although advances in the prediction and treatment of OHSS have been made, complete prevention has not been possible yet. S1P in follicular fluid from women at risk of developing OHSS are lower in comparison from women who are not at such risk and administration of S1P in an OHSS rat model decreases ovarian capillary permeability. STUDY DESIGN, SIZE, DURATION: We used an animal model that develops OHSS in immature Sprague-Dawley rats. The rats were randomly divided into three groups: the control group, which was injected with 10 IU of pregnant mare's serum gonadotropin (PMSG), and 10 IU of hCG 48 h later; the OHSS group, which was injected with excessive doses of PMSG (50 IU/day) for four consecutive days, followed by hCG; and the OHSS + S1P group, which was injected with the same doses of PMSG and hCG as the OHSS group and then treated with 5 mul S1P (1 mM) under the bursa of both ovaries, whereas the other groups of animals received the S1P vehicle. PARTICIPANTS /MATERIALS, SETTING, METHODS: Rats were killed by decapitation 48 h after the hCG injection for ovary, endometrium and blood collection. The ovaries were weighed and then used for subsequent assays, while the serum was used for hormone assays. One of the ovaries from each rat (n = 6) was used for Western immunoblot and the other for immunohistochemical analysis. Statistical comparisons between groups were carried out. MAIN RESULTS AND THE ROLE OF CHANCE: S1P administration reduced the ovarian weight (P < 0.05), and decreased the concentration of serum progesterone in the OHSS group compared to the OHSS group without treatment (P < 0.001). The percentage of antral follicles in the OHSS group was lower than that in the control group. S1P increased the percentage of antral follicles (P < 0.05) and decreased the percentage of corpora lutea (P < 0.01) and cystic structures in the OHSS group (P < 0.05). S1P had no effect on the expression levels of the enzymes 3beta-hydroxysteroid dehydrogenase (3betaHSD) or cholesterol side-chain cleavage enzyme (P450scc), but reduced the levels of steroidogenic acute regulatory protein (StAR) in OHSS rat ovaries (P < 0.05). S1P decreased the endothelial (P < 0.05) and periendothelial (P < 0.01) cell area in OHSS rat ovaries. S1P restored the levels of N-cadherin and VE-cadherin proteins to control values. Furthermore, S1P enhanced the levels of claudin-5, occludin (P < 0.05) and Sphingosine-1-phosphate receptor 1 (S1PR1) in OHSS (P < 0.01). In addition, no histological differences were found in endometrium between OHSS and S1P-treated OHSS animals. LIMITATIONS REASONS FOR CAUTION: The results of this study were generated from an in vivo OHSS experimental model, which has been used by several authors and our group due to the similarity between the rat and human angiogenic systems. Further studies in patients will be needed to evaluate the effects of S1P in the pathogenesis of OHSS. WIDER IMPLICATIONS OF THE FINDINGS: These findings concern the pathophysiological importance of S1P in OHSS. More studies on the regulation of endothelial cell barrier function by S1P in reproductive pathological processes and its therapeutic application are required. LARGE SCALE DATA: N/A. STUDY FUNDING AND COMPETING INTEREST(S): This work was supported by grants from ANPCyT (PICT 2012-897), CONICET (PIP 5471), Roemmers and Baron Foundations, Argentina. The authors declare no conflicts of interest.

Sphingosine 1-phosphate dynamically regulates myosin light chain phosphatase activity in human endothelial cells.[Pubmed:11955953]

Cell Signal. 2002 Jul;14(7):607-13.

Sphingosine 1-phosphate (S1P) is known to induce reorganization of the actin cytoskeleton through activation of the GTPase Rho. We have investigated the dynamic behavior of Rho/Rho kinase-regulated myosin light chain (MLC) phosphatase activity and MLC phosphorylation in Human Umbilical Vein Endothelial Cells (HUVEC) stimulated with S1P. Immediately (30-60 s) after S1P stimulation, MLC phosphatase activity dropped and MLC phosphorylation increased in a Rho/Rho kinase-dependent manner. Shortly thereafter (2 min), MLC phosphatase increased above baseline and MLC phosphorylation correspondingly decreased to near control values. At this time point, formation of actin ruffles and Rac activity assays indicated activation of Rac. Finally, between 5 and 15 min, MLC phosphatase dropped to a plateau below baseline. In parallel, MLC phosphorylation became constantly elevated above control values. These findings indicate that S1P is able to induce dynamic cycles of MLC phosphatase deactivation and activation. This novel feature of S1P could contribute to its chemotactic and angiogenic activity.

Sphingosine 1-phosphate is a ligand of the human gpr3, gpr6 and gpr12 family of constitutively active G protein-coupled receptors.[Pubmed:12220620]

Cell Signal. 2002 Nov;14(11):941-53.

Five G protein-coupled receptors (GPCRs) for the lysophospholipid sphingosine 1-phosphate (S1P) have been cloned and characterized so far. We report here about the identification of gpr3, gpr6 and gpr12 as additional members of the S1P-GPCR family. When expressed transiently in HEK293 cells, gpr3, gpr6 and gpr12 confer constitutive activation of adenylate cyclase (AC) similar in amplitude to that seen with fully activated G(alpha)(s)-coupled receptors. Culturing the transfected cells in medium with charcoal-stripped serum (devoid of lipids) significantly reduces cyclic adenosine monophosphate (cAMP) levels, suggesting a lipid-like ligand. A library containing 200 bioactive lipids was applied in functional assays recording intracellular Ca(2+) mobilization. S1P and dihydrosphingosine 1-phosphate (DHS1P) were identified as functional activators exhibiting nanomolar EC(50) values. In the presence of the S1P and LPA receptor antagonist suramin, gpr3-, gpr6- and gpr12-mediated intracellular Ca(2+) mobilization via S1P is enhanced. Besides constitutive activation of G(alpha)(s) type of G proteins, all three receptors are capable of constitutively activating inhibitory G(alpha)(i/o) proteins: (i) in the presence of pertussis toxin, gpr3-, gpr6- and gpr12-mediated stimulation of AC is enhanced; and (ii) overexpression of G(alpha)(i) significantly reduces the stimulatory action on intracellular cAMP levels. Agonist (S1P)-mediated internalization can be visualized in intact HEK293 cells using a gpr6 green fluorescent protein (GFP) fusion protein. In summary, our data suggest that gpr3, gpr6 and gpr12 are a family of constitutively active receptors with dual coupling to G(alpha)(s) and G(alpha)(i) type of G proteins. Constitutive activation of AC and mobilization of [Ca(2+)](i) can be modulated by the sphingophospholipids S1P and DHS1P, adding three additional members to the family of S1P receptors.

Sphingosine 1-phosphate signalling via the endothelial differentiation gene family of G-protein-coupled receptors.[Pubmed:11150592]

Pharmacol Ther. 2000 Nov;88(2):115-31.

Sphingosine 1-phosphate (S1P) is stored in and released from platelets in response to cell activation. However, recent studies show that it is also released from a number of cell types, where it can function as a paracrine/autocrine signal to regulate cell proliferation, differentiation, survival, and motility. This review discusses the role of S1P in cellular regulation, both at the molecular level and in terms of health and disease. The main biochemical routes for S1P synthesis (sphingosine kinase) and degradation (S1P lyase and S1P phosphatase) are described. The major focus is on the ability of S1P to bind to a novel family of G-protein-coupled receptors (endothelial differentiation gene [EDG]-1, -3, -5, -6, and -8) to elicit signal transduction (via G(q)-, G(i)-, G(12)-, G(13)-, and Rho-dependent routes). Effector pathways regulated by S1P are divergent, such as extracellular signal-regulated kinase, p38 mitogen-activated protein kinase, phospholipases C and D, adenylyl cyclase, and focal adhesion kinase, and occur in multiple cell types, such as immune cells, neurones, smooth muscle, etc. This provides a molecular basis for the ability of S1P to act as a pleiotropic bioactive lipid with an important role in cellular regulation. We also give an account of the expanding role for S1P in health and disease; in particular, with regard to its role in atherosclerosis, angiogenesis, cancer, and inflammation. Finally, we describe future directions for S1P research and novel approaches whereby S1P signalling can be manipulated for therapeutic intervention in disease.

Activation of a high affinity Gi protein-coupled plasma membrane receptor by sphingosine-1-phosphate.[Pubmed:8567663]

J Biol Chem. 1996 Jan 26;271(4):2082-7.

Sphingosine-1-phosphate (SPP) has attracted much attention as a possible second messenger controlling cell proliferation and motility and as an intracellular Ca(2+)-releasing agent. Here, we present evidence that SPP activates a G protein-coupled receptor in the plasma membrane of various cells, leading to increase in cytoplasmic Ca2+ concentration ([Ca2+]i), inhibition of adenylyl cyclase, and opening of G protein-regulated potassium channels. In human enbryonic kidney (HEK) cells, SPP potently (EC50, 2 nM) and rapidly increased [Ca2+]i in a pertussis toxin-sensitive manner. Pertussis toxin-sensitive increase in [Ca2+]i was also observed with sphingosylphosphorylcholine (EC50, 460 nM), whereas other sphingolipids, including ceramide-1-phosphate, N-palmitoyl-sphingosine, psychosine, and D-erythro-sphingosine at micromolar concentrations did not or only marginally increased [Ca2+]i. Furthermore, SPP inhibited forskolin-stimulated cAMP accumulation in HEK cells and increased binding of guanosine 5'3-O-(thio) triphosphate to HEK cell membranes. Rapid [Ca2+]i responses were also observed in human transitional bladder carcinoma (J82) cells, monkey COS-1 cells, mouse NIH 3T3 cells, Chinese hamster ovary (CHO-K1) cells, and rat C6 glioma cells, whereas human HL-60 leukemia cells and human erythroleukemia cells failed to respond to SPP. In guinea pig atrial myocytes, SPP activated Gi protein-regulated inwardly rectifying potassium channels. Activation of these channels occurred strictly when SPP was applied at the extracellular face of atrial myocyte plasma membrane as measured in cell-attached and inside-out patch clamp current recordings. We conclude that SPP, in addition to its proposed direct action on intracellular Ca2+ stores, interacts with a high affinity Gi protein-coupled receptor in the plasma membrane of apparently many different cell types.

Description

Endogenous agonist at S1P1-5

Keywords:

Sphingosine-1-phosphate,26993-30-6,Natural Products,Sphingosine, buy Sphingosine-1-phosphate , Sphingosine-1-phosphate supplier , purchase Sphingosine-1-phosphate , Sphingosine-1-phosphate cost , Sphingosine-1-phosphate manufacturer , order Sphingosine-1-phosphate , high purity Sphingosine-1-phosphate

Online Inquiry for:

      Fill out the information below

      • Size:Qty: - +

      * Required Fields

                                      Result: