801
|
Cinamon G, Matloubian M, Lesneski MJ, Xu Y, Low C, Lu T, Proia RL, Cyster JG. Sphingosine 1-phosphate receptor 1 promotes B cell localization in the splenic marginal zone. Nat Immunol 2004; 5:713-20. [PMID: 15184895 DOI: 10.1038/ni1083] [Citation(s) in RCA: 320] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2004] [Accepted: 04/13/2004] [Indexed: 11/08/2022]
Abstract
The factors directing marginal zone B cells to the splenic marginal zone are not well understood. Here we report that FTY720, a drug that targets sphingosine 1-phosphate (S1P) receptors, induced marginal zone B cell migration into follicles. Marginal zone B cells expressed S1P receptors 1 and 3 (S1P(1) and S1P(3), respectively). Using gene-targeted mice, we show that S1P(1) but not S1P(3) was required for localization in the marginal zone. In mice lacking the chemokine CXCL13, S1P(1)-deficient marginal zone B cells reacquired a marginal zone distribution. Exposure to lipopolysaccharide or antigen caused marginal zone B cells to downregulate S1P(1) and S1P(3) and to migrate into the splenic white pulp. These data suggest that marginal zone B cell localization to the marginal zone depends on responsiveness to the blood lysophospholipid S1P, with S1P(1) signaling overcoming the recruiting activity of CXCL13.
Collapse
Affiliation(s)
- Guy Cinamon
- Howard Hughes Medical Institute, Department of Microbiology, University of California San Francisco, 513 Parnassus Avenue, San Francisco, California 94143-0414, USA
| | | | | | | | | | | | | | | |
Collapse
|
802
|
Kostenis E. A glance at G-protein-coupled receptors for lipid mediators: a growing receptor family with remarkably diverse ligands. Pharmacol Ther 2004; 102:243-57. [PMID: 15246248 DOI: 10.1016/j.pharmthera.2004.04.005] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
A plethora of lipid-like molecules known to act as intracellular second messengers are now recognized to signal cells through plasma membrane 7 transmembrane G-protein-coupled receptors (GPCRs). This has been the result of a decade-long genetic hunt for novel sequences encoding 7 transmembrane receptor proteins and the efforts to pair novel sequences with biologically active substances of (partly) unknown molecular mechanism of action. Identification of novel GPCR ligand pairs represents the first step to shed more light into the mode of action of novel cellular signaling molecules in human health and disease and might represent a fruitful source for the development of new drugs, judged on the successful history of GPCR as drug targets. Since 2000, more than 16 reports became available on lipid mediators--as diverse as lysophospholipids, arachidonic acid metabolites, short-, medium-, and long-chain fatty acids as well as steroid-like molecules--exerting their effects as extracellular mediators via rhodopsin-like family GPCRs. These reports have opened new avenues for research in human lipid receptor physiology and pharmacology. Here, the current knowledge on the recently deorphanized lipid receptors, including their isolation, expression pattern, function, and possible physiological or pathological roles will be reviewed.
Collapse
Affiliation(s)
- Evi Kostenis
- 7TM Pharma A/S, 3 Fremtidsvej, 2970 Hoersholm, Denmark.
| |
Collapse
|
803
|
Waters C, Pyne S, Pyne NJ. The role of G-protein coupled receptors and associated proteins in receptor tyrosine kinase signal transduction. Semin Cell Dev Biol 2004; 15:309-23. [PMID: 15125894 DOI: 10.1016/j.semcdb.2003.12.020] [Citation(s) in RCA: 65] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
It is well established that stimulation of G-protein coupled receptors (GPCRs) can activate signalling from receptor tyrosine kinases by a process termed transactivation. Indeed, in recent years, it has become apparent that transactivation is a general phenomenon that has been demonstrated for many unrelated GPCRs and receptor tyrosine kinases. In this case the GPCR/G-protein participation is up-stream of the receptor tyrosine kinase. Substantial research has addressed these findings but meanwhile another mechanism of cross talk has been slowly emerging. For over a decade, a growing body of evidence has demonstrated that numerous growth factors use G-proteins and attendant signalling molecules such as beta-arrestins that participate down-stream of the receptor tyrosine kinase to signal to effectors, such as p42/p44 MAPK. This review highlights this novel mechanism of cross talk between receptor tyrosine kinases and GPCRs, which is distinct from growth factor receptor transactivation by GPCRs.
Collapse
Affiliation(s)
- Catherine Waters
- Department of Physiology and Pharmacology, Strathclyde Institute for Biomedical Sciences, University of Strathclyde, 27 Taylor Street, Glasgow G4 ONR, Scotland, UK
| | | | | |
Collapse
|
804
|
Kono M, Mi Y, Liu Y, Sasaki T, Allende ML, Wu YP, Yamashita T, Proia RL. The sphingosine-1-phosphate receptors S1P1, S1P2, and S1P3 function coordinately during embryonic angiogenesis. J Biol Chem 2004; 279:29367-73. [PMID: 15138255 DOI: 10.1074/jbc.m403937200] [Citation(s) in RCA: 328] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Sphingosine-1-phosphate (S1P) elicits diverse cellular responses through a family of G-protein-coupled receptors. We have shown previously that genetic disruption of the S1P(1) receptor, the most widely expressed of the family, results in embryonic lethality because of its key role within endothelial cells in regulating the coverage of blood vessels by vascular smooth muscle cells. To understand the physiologic functions of the two other widely expressed S1P receptors, we generated S1P(2) and S1P(3) null mice. Neither the S1P(2) null mice nor the S1P(3) null mice exhibited significant embryonic lethality or obvious phenotypic abnormalities. To unmask possible overlapping or collaborative functions between the S1P(1), S1P(2), and S1P(3) receptors, we examined embryos with multiple S1P receptor mutations. We found that S1P(1) S1P(2) double null and S1P(1) S1P(2) S1P(3) triple null embryos displayed a substantially more severe vascular phenotype than did embryos with only S1P(1) deleted. We also found partial embryonic lethality and vascular abnormalities in S1P(2) S1P(3) double null embryos. Our results indicate that the S1P(1), S1P(2) and S1P(3) receptors have redundant or cooperative functions for the development of a stable and mature vascular system during embryonic development.
Collapse
Affiliation(s)
- Mari Kono
- Genetics of Development and Disease Branch, NIDDK, National Institutes of Health, Bethesda, Maryland 20892, USA
| | | | | | | | | | | | | | | |
Collapse
|
805
|
Zhou H, Murthy KS. Distinctive G protein-dependent signaling in smooth muscle by sphingosine 1-phosphate receptors S1P1and S1P2. Am J Physiol Cell Physiol 2004; 286:C1130-8. [PMID: 15075212 DOI: 10.1152/ajpcell.00429.2003] [Citation(s) in RCA: 83] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
We examined expression of sphingosine 1-phosphate (S1P) receptors and sphingosine kinase (SPK) in gastric smooth muscle cells and characterized signaling pathways mediating S1P-induced 20-kDa myosin light chain (MLC20) phosphorylation and contraction. RT-PCR demonstrated expression of SPK1 and SPK2 and S1P1and S1P2receptors. S1P activated Gq, G13, and all Giisoforms and stimulated PLC-β1, PLC-β3, and Rho kinase activities. PLC-β activity was partially inhibited by pertussis toxin (PTX), Gβ or Gαqantibody, PLC-β1 or PLC-β3 antibody, and by expression of Gαqor Gαiminigene, and was abolished by a combination of antibodies or minigenes. S1P-stimulated Rho kinase activity was partially inhibited by expression of Gα13or Gαqminigene and abolished by expression of both. S1P stimulated Ca2+release that was inhibited by U-73122 and heparin and induced concentration-dependent contraction of smooth muscle cells (EC501 nM). Initial contraction and MLC20phosphorylation were abolished by U-73122 and MLC kinase (MLCK) inhibitor ML-9. Initial contraction was also partially inhibited by PTX and Gαqor Gβ antibody and abolished by a combination of both antibodies. In contrast, sustained contraction and MLC20phosphorylation were partially inhibited by a PKC or Rho kinase inhibitor (bisindolylmaleimide and Y-27632) and abolished by a combination of both inhibitors but not affected by U-73122 or ML-9. These results indicate that S1P induces 1) initial contraction mediated by S1P2and S1P1involving concurrent activation of PLC-β1 and PLC-β3 via Gαqand Gβγi, respectively, resulting in inositol 1,4,5-trisphosphate-dependent Ca2+release and MLCK-mediated MLC20phosphorylation, and 2) sustained contraction exclusively mediated by S1P2involving activation of RhoA via Gαqand Gα13, resulting in Rho kinase- and PKC-dependent MLC20phosphorylation.
Collapse
Affiliation(s)
- Huiping Zhou
- Depts. of Physiology and Medicine, Medical College of Virginia Campus, Virginia Commonwealth Univ., Richmond, VA 23298, USA
| | | |
Collapse
|
806
|
Forrest M, Sun SY, Hajdu R, Bergstrom J, Card D, Doherty G, Hale J, Keohane C, Meyers C, Milligan J, Mills S, Nomura N, Rosen H, Rosenbach M, Shei GJ, Singer II, Tian M, West S, White V, Xie J, Proia RL, Mandala S. Immune cell regulation and cardiovascular effects of sphingosine 1-phosphate receptor agonists in rodents are mediated via distinct receptor subtypes. J Pharmacol Exp Ther 2004; 309:758-68. [PMID: 14747617 DOI: 10.1124/jpet.103.062828] [Citation(s) in RCA: 285] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Sphingosine 1-phosphate (S1P) is a bioactive lysolipid with pleiotropic functions mediated through a family of G protein-coupled receptors, S1P(1,2,3,4,5). Physiological effects of S1P receptor agonists include regulation of cardiovascular function and immunosuppression via redistribution of lymphocytes from blood to secondary lymphoid organs. The phosphorylated metabolite of the immunosuppressant agent FTY720 (2-amino-2-(2-[4-octylphenyl]ethyl)-1,3-propanediol) and other phosphonate analogs with differential receptor selectivity were investigated. No significant species differences in compound potency or rank order of activity on receptors cloned from human, murine, and rat sources were observed. All synthetic analogs were high-affinity agonists on S1P(1), with IC(50) values for ligand binding between 0.3 and 14 nM. The correlation between S1P(1) receptor activation and the ED(50) for lymphocyte reduction was highly significant (p < 0.001) and lower for the other receptors. In contrast to S1P(1)-mediated effects on lymphocyte recirculation, three lines of evidence link S1P(3) receptor activity with acute toxicity and cardiovascular regulation: compound potency on S1P(3) correlated with toxicity and bradycardia; the shift in potency of phosphorylated-FTY720 for inducing lymphopenia versus bradycardia and hypertension was consistent with affinity for S1P(1) relative to S1P(3); and toxicity, bradycardia, and hypertension were absent in S1P(3)(-/-) mice. Blood pressure effects of agonists in anesthetized rats were complex, whereas hypertension was the predominant effect in conscious rats and mice. Immunolocalization of S1P(3) in rodent heart revealed abundant expression on myocytes and perivascular smooth muscle cells consistent with regulation of bradycardia and hypertension, whereas S1P(1) expression was restricted to the vascular endothelium.
Collapse
Affiliation(s)
- M Forrest
- Merck Research Laboratories, Department of Immunology and Rheumatology, Rahway, NJ 07065, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
807
|
Chen S, Lechleider RJ. Transforming growth factor-beta-induced differentiation of smooth muscle from a neural crest stem cell line. Circ Res 2004; 94:1195-202. [PMID: 15059931 DOI: 10.1161/01.res.0000126897.41658.81] [Citation(s) in RCA: 149] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
During vascular development, nascent endothelial networks are invested with a layer of supporting cells called pericytes in capillaries or smooth muscle in larger vessels. The cellular lineage of smooth muscle precursors and factors responsible for regulating their differentiation remain uncertain. In vivo, cells derived from the multipotent neural crest can give rise to vascular smooth muscle in parts of the head and also the cardiac outflow tract. Although transforming growth factor-beta (TGF-beta) has previously been shown to induce some smooth muscle markers from primary cultures of neural crest stem cells, the extent of the differentiation induced was not clear. In this study, we demonstrate that TGF-beta can induce many of the markers and characteristics of vascular smooth muscle from a neural crest stem cell line, Monc-1. Within 3 days of in vitro treatment, TGF-beta induces multiple smooth muscle-specific markers, while downregulating epithelial markers present on the parent cells. Treatment with TGF-beta also induces a contractile phenotype that responds to the muscarinic agonist carbachol and is not immediately reversed on TGF-beta withdrawal. Examination of the signaling pathways involved revealed that TGF-beta activation of Smad2 and Smad3 appear to be essential for the observed differentiation. Taken together, this system provides a novel model of smooth muscle differentiation that reliably recapitulates the process observed in vivo and allows for dissection of the pathways and processes involved in this process.
Collapse
Affiliation(s)
- Shiyou Chen
- Department of Cell Biology, Georgetown University Medical School, Washington, DC 20057, USA
| | | |
Collapse
|
808
|
Chae SS, Paik JH, Allende ML, Proia RL, Hla T. Regulation of limb development by the sphingosine 1-phosphate receptor S1p1/EDG-1 occurs via the hypoxia/VEGF axis. Dev Biol 2004; 268:441-7. [PMID: 15063179 DOI: 10.1016/j.ydbio.2004.01.001] [Citation(s) in RCA: 58] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2003] [Revised: 12/07/2003] [Accepted: 01/05/2004] [Indexed: 11/15/2022]
Abstract
Angiogenesis, also known as new blood vessel formation, is regulated coordinately with other tissue differentiation events during limb development. Although vascular endothelial cell growth factor (VEGF) is important in the regulation of angiogenesis, chondrogenesis and osteogenesis during limb development, the role of other angiogenic factors is not well understood. Sphingosine 1-phosphate, a platelet-derived lipid mediator, regulates angiogenesis and vascular maturation via its action on the G-protein-coupled receptor S1P(1) (also known as EDG-1). In addition to vascular defects, abnormal limb development was also observed in S1p(1)(-/-) mice. Here we show that strong induction of S1P(1) expression is observed in the blood vessels and the interdigital mesenchymal cells during limb development. Deletion of S1P(1) results in aberrant chondrocyte condensation and defective digit morphogenesis. Interestingly, the vasculature in the S1p(1)(-/-) limbs was hyperplastic and morphologically altered. In addition, the hypoxia inducible factor (HIF)-1 alpha and its response gene VEGF were induced in S1p(1)(-/-) limbs. However, aberrant regulation of HIF-1 alpha and VEGF were not observed in embryonic fibroblasts derived from S1p(1)(-/-) mice, suggesting a non-cell autonomous effect of S1P(1) on VEGF expression. Indeed, similar limb defects were observed in endothelium-specific S1P(1) null mice in vivo. These data suggest that the function of S1P(1) in the developing vasculature is essential for proper limb development.
Collapse
Affiliation(s)
- Sung-Suk Chae
- Center for Vascular Biology, Department of Cell Biology, University of Connecticut Health Center, Farmington, CT 06030-3501, USA
| | | | | | | | | |
Collapse
|
809
|
Mo FM, Offertáler L, Kunos G. Atypical cannabinoid stimulates endothelial cell migration via a Gi/Go-coupled receptor distinct from CB1, CB2 or EDG-1. Eur J Pharmacol 2004; 489:21-7. [PMID: 15063151 DOI: 10.1016/j.ejphar.2004.02.034] [Citation(s) in RCA: 54] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2004] [Accepted: 02/26/2004] [Indexed: 11/30/2022]
Abstract
The endothelium-dependent mesenteric vasorelaxant effect of anandamide has been attributed to stimulation of a Gi/Go-coupled receptor, for which the nonpsychoactive analog abnormal cannabidiol (abn-cbd, (-)-4-(3-3,4-trans-p-menthadien-[1,8]-yl)olivetol) is a selective agonist and the compound O-1918 ((-)-4-(3-3,4-trans-p-menthadien-(1,8)-yl)-orcinol) is a selective antagonist. In human umbilical vein endothelial cells abn-cbd was reported to increase the phosphorylation of p44/42 mitogen activated protein kinase (MAPK) and protein kinase B/Akt, and these effects could be inhibited by pertussis toxin, by phosphatidylinositol 3-kinase (PI3K) inhibitors or by O-1918 [Mol. Pharmacol. 63 (2003) 699]. In the present experiments, abn-cbd caused a concentration-dependent increase in human umbilical vein endothelial cell migration, as quantified in a transwell chamber. This effect was antagonized by O-1918, by the PI3K inhibitor wortmannin, and by pertussis toxin, but not by the cannabinoid CB1 receptor antagonist AM251 or the cannabinoid CB2 receptor antagonist SR144528. The EDG-1 receptor agonist sphingosine-1-phosphate also increased human umbilical vein endothelial cell migration, but this response was unaffected by O-1918. In Chinese hamster ovary cells stably transfected with the gene encoding the EDG-1 receptor, p44/42 MAPK phosphorylation was unaffected by abn-cbd, but strongly induced by sphingosine-1-phosphate. These results indicate that an abn-cbd-sensitive endothelial receptor distinct from cannabinoid CB1, CB2 or EDG-1 receptors mediates not only vasorelaxation but also endothelial cell migration.
Collapse
MESH Headings
- Androstadienes/pharmacology
- Animals
- Blotting, Western
- CHO Cells
- Cannabidiol/analogs & derivatives
- Cannabidiol/pharmacology
- Cell Movement/drug effects
- Cells, Cultured
- Cricetinae
- Endothelial Cells/drug effects
- Enzyme Inhibitors/pharmacology
- GTP-Binding Protein alpha Subunits, Gi-Go/drug effects
- Humans
- Indicators and Reagents
- Lysophospholipids/pharmacology
- Mitogen-Activated Protein Kinase 1/antagonists & inhibitors
- Mitogen-Activated Protein Kinase 1/metabolism
- Pertussis Toxin/pharmacology
- Receptor, Cannabinoid, CB1/antagonists & inhibitors
- Receptor, Cannabinoid, CB1/drug effects
- Receptor, Cannabinoid, CB2/antagonists & inhibitors
- Receptor, Cannabinoid, CB2/drug effects
- Receptors, Estrogen/drug effects
- Receptors, G-Protein-Coupled/drug effects
- Receptors, Lysosphingolipid
- Resorcinols/pharmacology
- Sphingosine/analogs & derivatives
- Sphingosine/pharmacology
- Sphingosine-1-Phosphate Receptors
- Umbilical Veins/cytology
- Wortmannin
Collapse
Affiliation(s)
- Fong Ming Mo
- Laboratory of Physiologic Studies, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD 20892-8115, USA
| | | | | |
Collapse
|
810
|
Abbey-Hosch SE, Cody AN, Potter LR. Sphingosine-1-phosphate inhibits C-type natriuretic peptide activation of guanylyl cyclase B (GC-B/NPR-B). Hypertension 2004; 43:1103-9. [PMID: 15037564 DOI: 10.1161/01.hyp.0000124668.80811.d3] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
C-type natriuretic peptide (CNP) binds and activates the transmembrane guanylyl cyclase B receptor (NPR-B), which decreases vascular tone and inhibits cell proliferation and migration. In contrast, the bioactive lipid sphingosine-1-phosphate (S1P) elicits the opposite physiological effects. Here, we demonstrate a potent acute inhibitory effect of S1P on NPR-B activity in NIH3T3 fibroblasts and A10 vascular smooth muscle cells. In fibroblasts, S1P reduced CNP-dependent cGMP elevations to the same levels as 10% fetal bovine serum, the most potent NPR-B desensitizing agent known. The reduction was dose-dependent (IC50=0.08 micromol/L) and due to decreased NPR-B activity because CNP-dependent guanylyl cyclase activities were markedly diminished in membranes prepared from S1P-treated cells. Similarly, in A10 cells, S1P inhibition was rapid (t1/2=2 to 5 minutes), dose-dependent (IC50=0.3 micromol/L S1P), and mediated by a cell surface receptor. The mechanism of the S1P-dependent desensitization in A10 cells did not require NPR-B degradation or protein kinase C activation, but did require elevated calcium concentrations because a nonspecific calcium ionophore also inhibited NPR-B and an intracellular calcium chelator blocked a significant portion of the S1P response. These are the first data demonstrating cross-talk between the natriuretic peptide and S1P signaling systems. They suggest that the effects of S1P on vascular disease and wound healing may be mediated in part through inhibition of NPR-B.
Collapse
MESH Headings
- Animals
- Cell Division/drug effects
- Cyclic GMP/metabolism
- Dose-Response Relationship, Drug
- Egtazic Acid/analogs & derivatives
- Egtazic Acid/pharmacology
- Enzyme Activation/drug effects
- Enzyme Inhibitors/pharmacology
- Fibroblasts/drug effects
- Fibroblasts/enzymology
- Guanylate Cyclase/antagonists & inhibitors
- Indoles/pharmacology
- Inhibitory Concentration 50
- Ionomycin/pharmacology
- Maleimides/pharmacology
- Mice
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/enzymology
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/enzymology
- NIH 3T3 Cells/drug effects
- NIH 3T3 Cells/enzymology
- Natriuretic Peptide, C-Type/administration & dosage
- Natriuretic Peptide, C-Type/pharmacology
- Organ Specificity
- Rats
- Receptors, Cell Surface/antagonists & inhibitors
- Receptors, Cell Surface/physiology
- Signal Transduction/drug effects
- Signal Transduction/physiology
- Sphingosine/administration & dosage
- Sphingosine/analogs & derivatives
- Sphingosine/pharmacology
Collapse
Affiliation(s)
- Sarah E Abbey-Hosch
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis, MN 55455, USA
| | | | | |
Collapse
|
811
|
Abstract
The many biological responses documented for lysophospholipids that include lysophosphatidic acid and sphingosine 1-phosphate can be mechanistically attributed to signaling through specific G protein-coupled receptors. At least nine receptors have now been identified, and the total number is likely to be larger. In this brief review, we note cogent features of lysophospholipid receptors, including the current nomenclature, signaling properties, development of agonists and antagonists, and physiological functions.
Collapse
Affiliation(s)
- Brigitte Anliker
- Department of Molecular Biology, Helen L. Dorris Institute for Neurological and Psychiatric Disorders, The Scripps Research Institute, La Jolla, CA 92037, USA
| | | |
Collapse
|
812
|
Harada J, Foley M, Moskowitz MA, Waeber C. Sphingosine-1-phosphate induces proliferation and morphological changes of neural progenitor cells. J Neurochem 2004; 88:1026-39. [PMID: 14756825 DOI: 10.1046/j.1471-4159.2003.02219.x] [Citation(s) in RCA: 161] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Sphingosine-1-phosphate (S1P) is a lipid mediator that exerts multiple cellular functions through activation of G-protein-coupled receptors. Although the role of S1P on angiogenesis is well established, its role in neurogenesis is unknown. We examined the effects of S1P on G-protein activation in brain sections of rat embryo and on neural progenitor cells in culture. Intense S1P-stimulated [35S]GTPgammaS labeling was observed as early as E15 in the neuroepithelium and differentiating fields throughout the brain, suggesting that functional S1P receptors are expressed in brain areas with active neurogenesis. mRNA transcripts for several S1P receptor subtypes (S1P1, S1P2, S1P3 and S1P5) were expressed in neural progenitor cells prepared from embryonic rat hippocampus. S1P induced phosphorylation of extracellular signal-regulated kinase (ERK) and proliferation of neural progenitor cells as determined by BrdU incorporation in a pertussis toxin-sensitive manner. These effects were prevented by the ERK signaling inhibitor U0126. S1P augmented telomerase activity in neural progenitor cells with similar potency as that of FGF-2. Furthermore, S1P induced cell-cell aggregation. This morphological change was transient and prevented by Y-27632, an inhibitor of Rho-associated kinase. These results suggest that S1P plays a pleiotropic role in neurogenesis via pathways involving S1P receptors, MAP kinases and Rho kinase.
Collapse
Affiliation(s)
- Jun Harada
- Stroke and Neurovascular Regulation Laboratory, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Charlestown, Massachusetts 02129, USA
| | | | | | | |
Collapse
|
813
|
Matloubian M, Lo CG, Cinamon G, Lesneski MJ, Xu Y, Brinkmann V, Allende ML, Proia RL, Cyster JG. Lymphocyte egress from thymus and peripheral lymphoid organs is dependent on S1P receptor 1. Nature 2004; 427:355-60. [PMID: 14737169 DOI: 10.1038/nature02284] [Citation(s) in RCA: 2048] [Impact Index Per Article: 97.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2003] [Accepted: 12/12/2003] [Indexed: 12/18/2022]
Abstract
Adaptive immunity depends on T-cell exit from the thymus and T and B cells travelling between secondary lymphoid organs to survey for antigens. After activation in lymphoid organs, T cells must again return to circulation to reach sites of infection; however, the mechanisms regulating lymphoid organ exit are unknown. An immunosuppressant drug, FTY720, inhibits lymphocyte emigration from lymphoid organs, and phosphorylated FTY720 binds and activates four of the five known sphingosine-1-phosphate (S1P) receptors. However, the role of S1P receptors in normal immune cell trafficking is unclear. Here we show that in mice whose haematopoietic cells lack a single S1P receptor (S1P1; also known as Edg1) there are no T cells in the periphery because mature T cells are unable to exit the thymus. Although B cells are present in peripheral lymphoid organs, they are severely deficient in blood and lymph. Adoptive cell transfer experiments establish an intrinsic requirement for S1P1 in T and B cells for lymphoid organ egress. Furthermore, S1P1-dependent chemotactic responsiveness is strongly upregulated in T-cell development before exit from the thymus, whereas S1P1 is downregulated during peripheral lymphocyte activation, and this is associated with retention in lymphoid organs. We find that FTY720 treatment downregulates S1P1, creating a temporary pharmacological S1P1-null state in lymphocytes, providing an explanation for the mechanism of FTY720-induced lymphocyte sequestration. These findings establish that S1P1 is essential for lymphocyte recirculation and that it regulates egress from both thymus and peripheral lymphoid organs.
Collapse
Affiliation(s)
- Mehrdad Matloubian
- Howard Hughes Medical Institute and Department of Microbiology, University of California San Francisco, 513 Parnassus Avenue, San Francisco, California 94143-0414, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
814
|
Xia L, Ju T, Westmuckett A, An G, Ivanciu L, McDaniel JM, Lupu F, Cummings RD, McEver RP. Defective angiogenesis and fatal embryonic hemorrhage in mice lacking core 1-derived O-glycans. ACTA ACUST UNITED AC 2004; 164:451-9. [PMID: 14745002 PMCID: PMC2172228 DOI: 10.1083/jcb.200311112] [Citation(s) in RCA: 150] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The core 1 β1-3-galactosyltransferase (T-synthase) transfers Gal from UDP-Gal to GalNAcα1-Ser/Thr (Tn antigen) to form the core 1 O-glycan Galβ1-3GalNAcα1-Ser/Thr (T antigen). The T antigen is a precursor for extended and branched O-glycans of largely unknown function. We found that wild-type mice expressed the NeuAcα2-3Galβ1-3GalNAcα1-Ser/Thr primarily in endothelial, hematopoietic, and epithelial cells during development. Gene-targeted mice lacking T-synthase instead expressed the nonsialylated Tn antigen in these cells and developed brain hemorrhage that was uniformly fatal by embryonic day 14. T-synthase–deficient brains formed a chaotic microvascular network with distorted capillary lumens and defective association of endothelial cells with pericytes and extracellular matrix. These data reveal an unexpected requirement for core 1–derived O-glycans during angiogenesis.
Collapse
MESH Headings
- Animals
- Antigens, Tumor-Associated, Carbohydrate/genetics
- Antigens, Tumor-Associated, Carbohydrate/metabolism
- Antigens, Viral, Tumor/chemistry
- Antigens, Viral, Tumor/metabolism
- Blood Coagulation/physiology
- Embryo, Mammalian/anatomy & histology
- Embryo, Mammalian/pathology
- Embryo, Mammalian/physiology
- Endothelial Cells/metabolism
- Extracellular Matrix
- Female
- Galactosyltransferases/genetics
- Galactosyltransferases/metabolism
- Gestational Age
- Glycosylation
- Hemorrhage
- Humans
- Mice
- Mice, Knockout
- Microcirculation/anatomy & histology
- Microcirculation/metabolism
- Neovascularization, Physiologic
- Pericytes/metabolism
- Polysaccharides/chemistry
- Polysaccharides/metabolism
- Pregnancy
- Tissue Distribution
Collapse
Affiliation(s)
- Lijun Xia
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, 825 N.E. 13th St., Oklahoma City, OK 73104, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
815
|
Sanna MG, Liao J, Jo E, Alfonso C, Ahn MY, Peterson MS, Webb B, Lefebvre S, Chun J, Gray N, Rosen H. Sphingosine 1-phosphate (S1P) receptor subtypes S1P1 and S1P3, respectively, regulate lymphocyte recirculation and heart rate. J Biol Chem 2004; 279:13839-48. [PMID: 14732717 DOI: 10.1074/jbc.m311743200] [Citation(s) in RCA: 527] [Impact Index Per Article: 25.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Sphingosine 1-phosphate (S1P) influences heart rate, coronary artery caliber, endothelial integrity, and lymphocyte recirculation through five related high affinity G-protein-coupled receptors. Inhibition of lymphocyte recirculation by non-selective S1P receptor agonists produces clinical immunosuppression preventing transplant rejection but is associated with transient bradycardia. Understanding the contribution of individual receptors has been limited by the embryonic lethality of the S1P(1) knock-out and the unavailability of selective agonists or antagonists. A potent, S1P(1)-receptor selective agonist structurally unrelated to S1P was found to activate multiple signals triggered by S1P, including guanosine 5'-3-O-(thio)triphosphate binding, calcium flux, Akt and ERK1/2 phosphorylation, and stimulation of migration of S1P(1)- but not S1P(3)-expressing cells in vitro. The agonist also alters lymphocyte trafficking in vivo. Use of selective agonism together with deletant mice lacking S1P(3) receptor reveals that agonism of S1P(1) receptor alone is sufficient to control lymphocyte recirculation. Moreover, S1P(1) receptor agonist plasma levels are causally associated with induction and maintenance of lymphopenia. S1P(3), and not S1P(1), is directly implicated in sinus bradycardia. The sustained bradycardia induced by S1P receptor non-selective immunosuppressive agonists in wild-type mice is abolished in S1P(3)-/- mice, whereas S1P(1)-selective agonist does not produce bradycardia. Separation of receptor subtype usage for control of lymphocyte recirculation and heart rate may allow the identification of selective immunosuppressive S1P(1) receptor agonists with an enhanced therapeutic window. S1P(1)-selective agonists will be of broad utility in understanding cell functions in vitro, and vascular physiology in vivo, and the success of the chemical approach for S1P(1) suggests that selective tools for the resolution of function across this broad lipid receptor family are now possible.
Collapse
Affiliation(s)
- M Germana Sanna
- Department of Immunology, The Center for Mass Spectrometry, The Scripps Research Institute, La Jolla, California 92037, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
816
|
Allende ML, Dreier JL, Mandala S, Proia RL. Expression of the sphingosine 1-phosphate receptor, S1P1, on T-cells controls thymic emigration. J Biol Chem 2004; 279:15396-401. [PMID: 14732704 DOI: 10.1074/jbc.m314291200] [Citation(s) in RCA: 363] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
S1P(1) is a widely distributed G protein-coupled receptor whose ligand, sphingosine 1-phosphate, is present in high concentrations in the blood. The sphingosine 1-phosphate receptor-signaling pathway is believed to have potent effects on cell trafficking in the immune system. To determine the precise role of the S1P(1) receptor on T-cells, we established a T-cell-specific S1P(1) knock-out mouse. The mutant mice showed a block in the egress of mature T-cells into the periphery. The expression of the S1P(1) receptor was up-regulated in mature thymocytes, and its deletion altered the chemotactic responses of thymocytes to sphingosine 1-phosphate. The results indicated that the expression of the S1P(1) receptor on T-cells controls their exit from the thymus and entry into the blood and, thus, has a central role in regulating the numbers of peripheral T-cells.
Collapse
Affiliation(s)
- Maria L Allende
- Genetics of Development and Disease Branch, NIDDK, National Institutes of Health, Bethesda, Maryland 20892-1821, USA
| | | | | | | |
Collapse
|
817
|
Herr DR, Fyrst H, Creason MB, Phan VH, Saba JD, Harris GL. Characterization of the Drosophila sphingosine kinases and requirement for Sk2 in normal reproductive function. J Biol Chem 2004; 279:12685-94. [PMID: 14722126 DOI: 10.1074/jbc.m310647200] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Sphingosine kinase is a highly conserved enzyme that catalyzes the synthesis of sphingosine 1-phosphate and reduces cellular levels of sphingosine and ceramide. Although ceramide is pro-apoptotic and sphingosine is generally growth-inhibitory, sphingosine 1-phosphate signaling promotes cell proliferation, survival, and migration. Sphingosine kinase is thus in a strategic position to regulate important cell fate decisions which may contribute to normal animal development. To facilitate studies examining the potential role of sphingosine kinase and long chain base metabolism in Drosophila development, we characterized two putative Drosophila sphingosine kinase genes, Sk1 and Sk2. Both genes functionally and biochemically complement a yeast sphingosine kinase mutant, express predominantly cytosolic activities, and are capable of phosphorylating a range of endogenous and non-endogenous sphingoid base substrates. The two genes demonstrate overlapping but distinct temporal and spatial expression patterns in the Drosophila embryo, and timing of expression is consistent with observed changes in long chain base levels throughout development. A null Sk2 transposon insertion mutant demonstrated elevated long chain base levels, impaired flight performance, and diminished ovulation. This is the first reported mutation of a sphingosine kinase in an animal model; the associated phenotypes indicate that Sk1 and Sk2 are not redundant in biological function and that sphingosine kinase is essential for diverse physiological functions in this organism.
Collapse
Affiliation(s)
- Deron R Herr
- Department of Biology and Molecular Biology Institute, San Diego State University, San Diego, CA 92182-4614, USA
| | | | | | | | | | | |
Collapse
|
818
|
Usui S, Sugimoto N, Takuwa N, Sakagami S, Takata S, Kaneko S, Takuwa Y. Blood lipid mediator sphingosine 1-phosphate potently stimulates platelet-derived growth factor-A and -B chain expression through S1P1-Gi-Ras-MAPK-dependent induction of Kruppel-like factor 5. J Biol Chem 2004; 279:12300-11. [PMID: 14711826 DOI: 10.1074/jbc.m305025200] [Citation(s) in RCA: 84] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Platelet-derived growth factors (PDGFs), potent mitogens and chemoattractants for mesenchymal cell types, play essential roles in development of several organs including blood vessels, kidney, and lung, and are also implicated in the pathogenesis of atherosclerosis and malignancies. Blood lipid mediator sphingosine 1-phosphate (S1P) regulates migration, proliferation, and apoptosis in a variety of cell types through multiple G protein-coupled receptors of the Edg family, and is necessary for vascular formation at the developmental stage. We found in the present study that S1P induced severalfold increases in the mRNA and protein levels of PDGF-A and -B chains in vascular smooth muscle cells and neointimal cells. S1P stimulation of PDGF mRNA and protein expression was abolished by the small interfering RNA duplexes targeting S1P(1)/Edg1 receptor subtype. S1P stimulated the small GTPase Ras in a G(i)-dependent manner, and activated ERK and p38 MAPK in G(i)- and Ras-dependent manners. Pertussis toxin pretreatment, adenovirus-mediated Asn(17)Ras expression, the MEK inhibitor PD98059, or the p38 MAPK inhibitor SB203580 markedly suppressed PDGF mRNA and protein up-regulation, indicating the involvement of G(i)-Ras-ERK/p38 MAPK in S1P stimulation of PDGF expression. S1P stimulated expression of the transcription factor KLF5 in manners dependent on G(i), Ras, and ERK/p38 MAPK. Down-regulation of KLF5 by small interfering RNA duplexes abolished S1P-induced PDGF-A and -B chain expression. On the other hand, overexpression of KLF5 stimulated basal and S1P-induced PDGF expression. Either S1P stimulation or KLF5 overexpression increased the PDGF-B promoter activity in a cis-element-dependent manner. These results reveal the S1P(1)-triggered, G(i)-Ras-ERK/p38 MAPK-KLF5-dependent, stimulatory regulation of PDGF gene transcription in vascular smooth muscle cells.
Collapse
MESH Headings
- Adenoviridae/genetics
- Animals
- Animals, Newborn
- Aorta/pathology
- Apoptosis
- Blotting, Northern
- Blotting, Western
- Cell Division
- Cell Movement
- Down-Regulation
- Enzyme Inhibitors/pharmacology
- Flavonoids/pharmacology
- Gene Transfer Techniques
- Kruppel-Like Transcription Factors
- Lipid Metabolism
- Lysophospholipids/metabolism
- MAP Kinase Signaling System
- Male
- Mitogen-Activated Protein Kinases/metabolism
- Models, Biological
- Muscle, Smooth, Vascular/cytology
- Myocytes, Smooth Muscle/cytology
- Plasmids/metabolism
- Platelet-Derived Growth Factor/metabolism
- Promoter Regions, Genetic
- Proto-Oncogene Proteins c-sis/metabolism
- RNA Interference
- RNA, Messenger/metabolism
- Rats
- Rats, Wistar
- Reverse Transcriptase Polymerase Chain Reaction
- Sphingosine/analogs & derivatives
- Sphingosine/metabolism
- Time Factors
- Trans-Activators/chemistry
- Transcription, Genetic
- Up-Regulation
- p38 Mitogen-Activated Protein Kinases
Collapse
Affiliation(s)
- Soichiro Usui
- Department of Molecular and Cellular Physiology, Kanazawa University Graduate School of Medical Sciences, 13-1 Takara-machi, Kanazawa, Ishikawa 920-8640, Japan
| | | | | | | | | | | | | |
Collapse
|
819
|
Fyrst H, Herr DR, Harris GL, Saba JD. Characterization of free endogenous C14 and C16 sphingoid bases from Drosophila melanogaster. J Lipid Res 2004; 45:54-62. [PMID: 13130120 DOI: 10.1194/jlr.m300005-jlr200] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Sphingolipid metabolites function as signaling molecules in mammalian cells, influencing cell proliferation, migration, and death. Recently, sphingolipid signaling has been implicated in the regulation of developmental processes in Drosophila melanogaster. However, biochemical analysis of endogenous Drosophila sphingoid bases has not been reported. In this study, a rapid HPLC-based method was developed for the analysis of free sphingoid bases endogenous to Drosophila. Four molecular species of endogenous free sphingoid bases were observed in adult flies and identified as C14 and C16 sphingosine (Sph) and C14 and C16 dihydrosphingosine (DHS). The C14 molecular species were the most prevalent, accounting for approximately 94% of the total free sphingoid bases in adult wild-type flies. An Sph kinase (SK) mutant demonstrated significant accumulation of all four sphingoid bases, whereas a serine palmitoyltransferase mutant demonstrated low but detectable levels. When endogenous sphingoid bases were evaluated at different stages of development, the observed ratio of Sph to DHS increased significantly from early embryo to adulthood. Throughout development, this ratio was significantly lower in the SK mutant as compared with the wild-type. This is the first report describing analysis of free C14 and C16 sphingoid bases from Drosophila. The biochemical characterization of these lipids from mutant models of sphingolipid metabolism should greatly facilitate the analysis of the biological significance of these signaling molecules.
Collapse
Affiliation(s)
- Henrik Fyrst
- Children's Hospital, Oakland Research Institute, 5700 Martin Luther King Jr Way, Oakland, CA 94609, USA
| | | | | | | |
Collapse
|
820
|
Chang SH, Liu CH, Conway R, Han DK, Nithipatikom K, Trifan OC, Lane TF, Hla T. Role of prostaglandin E2-dependent angiogenic switch in cyclooxygenase 2-induced breast cancer progression. Proc Natl Acad Sci U S A 2003; 101:591-6. [PMID: 14688410 PMCID: PMC327192 DOI: 10.1073/pnas.2535911100] [Citation(s) in RCA: 304] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Overexpression of human cyclooxygenase 2 (COX-2) in the mammary glands of transgenic mice induces tissue-specific tumorigenic transformation. However, the molecular mechanisms involved are not yet defined. Here we show that COX-2 expressed in the epithelial cell compartment regulates angiogenesis in the stromal tissues of the mammary gland. Microvessel density increased before visible tumor growth and exponentially during tumor progression. Inhibition of prostanoid synthesis with indomethacin strongly decreased microvessel density and inhibited tumor progression. Up-regulation of angiogenic regulatory genes in COX-2 transgenic mammary tissue was also potently inhibited by indomethacin treatment, suggesting that prostanoids released from COX-2-expressing mammary epithelial cells induce angiogenesis. G protein-coupled receptors for the major product, prostaglandin E(2) (PGE(2)) EP(1-4), are expressed during mammary gland development, and EP(1,2,4) receptors were up-regulated in tumor tissue. PGE(2) stimulated the expression angiogenic regulatory genes in mammary tumor cells isolated from COX-2 transgenic mice. Such cells are tumorigenic in nude mice; however, treatment with Celecoxib, a COX-2-specific inhibitor, reduced tumor growth and microvessel density. These results define COX-2-derived PGE(2) as a potent inducer of angiogenic switch during mammary cancer progression.
Collapse
Affiliation(s)
- Sung-Hee Chang
- Center for Vascular Biology, Department of Cell Biology, University of Connecticut Health Center, Farmington, CT 06030-3501, USA
| | | | | | | | | | | | | | | |
Collapse
|
821
|
Soncin F, Mattot V, Lionneton F, Spruyt N, Lepretre F, Begue A, Stehelin D. VE-statin, an endothelial repressor of smooth muscle cell migration. EMBO J 2003; 22:5700-11. [PMID: 14592969 PMCID: PMC275406 DOI: 10.1093/emboj/cdg549] [Citation(s) in RCA: 105] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
The recruitment and proliferation of smooth muscle cells and pericytes are two key events for the stabilization of newly formed capillaries during angiogenesis and, when out of control in the adult, are the main causes of arteriosclerosis. We have identified a novel gene, named VE-statin for vascular endothelial-statin, which is expressed specifically by endothelial cells of the developing mouse embryo and in the adult, and in early endothelial progenitors. The mouse and human VE-statin genes have been located on chromosome 2 and 9, respectively, they span >10 kbp and are transcribed in two major variants arising from independent initiation sites. The VE-statin transcripts code for a unique protein of 30 kDa that contains a signal peptide and two epidermal growth factor (EGF)-like modules. VE-statin is found in the cellular endoplasmic reticulum and secreted in the cell supernatant. Secreted VE-statin inhibits platelet-derived growth factor (PDGF)-BB-induced smooth muscle cell migration, but has no effects on endothelial cell migration. VE-statin is the first identified inhibitor of mural cell migration specifically produced by endothelial cells.
Collapse
MESH Headings
- Amino Acid Sequence
- Animals
- Base Sequence
- Calcium-Binding Proteins
- Cell Division
- Cell Line
- Cell Movement
- Cells, Cultured
- Chromosomes, Human, Pair 9/genetics
- Cloning, Molecular
- DNA, Complementary/genetics
- DNA-Binding Proteins
- EGF Family of Proteins
- Endothelial Growth Factors/genetics
- Endothelial Growth Factors/physiology
- Endothelium, Vascular/growth & development
- Endothelium, Vascular/physiology
- Growth Inhibitors/genetics
- Growth Inhibitors/physiology
- Humans
- In Situ Hybridization, Fluorescence
- In Vitro Techniques
- Kruppel-Like Transcription Factors
- Mice
- Molecular Sequence Data
- Muscle, Smooth, Vascular/cytology
- Muscle, Smooth, Vascular/growth & development
- Muscle, Smooth, Vascular/physiology
- Neovascularization, Physiologic
- Proteins/genetics
- Proteins/physiology
- Transcription Factors/genetics
- Transcription Factors/physiology
Collapse
Affiliation(s)
- Fabrice Soncin
- CNRS UMR 8526, Institut de Biologie de Lille, 1 rue Calmette, 59021 Lille Cedex, France.
| | | | | | | | | | | | | |
Collapse
|
822
|
Langlois S, Gingras D, Béliveau R. Membrane type 1-matrix metalloproteinase (MT1-MMP) cooperates with sphingosine 1-phosphate to induce endothelial cell migration and morphogenic differentiation. Blood 2003; 103:3020-8. [PMID: 15070679 DOI: 10.1182/blood-2003-08-2968] [Citation(s) in RCA: 77] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Membrane type 1-matrix metalloproteinase (MT1-MMP) has been suggested to play an important role in angiogenesis, but the mechanisms involved remain incompletely understood. Using an in vitro model of angiogenesis in which cell migration of bovine aortic endothelial cells (BAECs) and their morphogenic differentiation into capillary-like structures on Matrigel are induced by overexpression of MT1-MMP, we show that the platelet-derived bioactive lipid sphingosine 1-phosphate (S1P) is the predominant serum factor essential for MT1-MMP-dependent migration and morphogenic differentiation activities. In the presence of S1P, MT1-MMP-dependent cell migration and morphogenic differentiation were inhibited by pertussis toxin, suggesting the involvement of Gi-protein-coupled receptor-mediated signaling. Accordingly, cotransfection of BAECs with MT1-MMP and a constitutively active Galphai2 (Q205L) mutant increased cell migration and morphogenic differentiation, whereas treatment of BAECs overexpressing MT1-MMP with antisense oligonucleotides directed against S1P1 and S1P3, the predominant S1P receptors, significantly inhibited both processes. These results demonstrate that MT1-MMP-induced migration and morphogenic differentiation involve the cooperation of the enzyme with platelet-derived bioactive lipids through S1P-mediated activation of Galphai-coupled S1P1 and S1P3 receptors. Given the important contribution of platelets to tumor angiogenesis, the stimulation of endothelial MT1-MMP function by S1P may thus constitute an important molecular event linking hemostasis to angiogenesis.
Collapse
MESH Headings
- Animals
- Cattle
- Cell Differentiation/drug effects
- Cell Movement/drug effects
- Cells, Cultured
- Endothelium, Vascular/cytology
- Endothelium, Vascular/drug effects
- Endothelium, Vascular/metabolism
- Humans
- Lysophospholipids/metabolism
- Lysophospholipids/pharmacology
- Matrix Metalloproteinases, Membrane-Associated
- Metalloendopeptidases/antagonists & inhibitors
- Metalloendopeptidases/genetics
- Metalloendopeptidases/metabolism
- Neovascularization, Physiologic
- Oligodeoxyribonucleotides, Antisense/genetics
- Oligodeoxyribonucleotides, Antisense/pharmacology
- Receptors, G-Protein-Coupled/antagonists & inhibitors
- Receptors, G-Protein-Coupled/genetics
- Receptors, G-Protein-Coupled/metabolism
- Receptors, Lysophospholipid
- Sphingosine/analogs & derivatives
- Sphingosine/metabolism
- Sphingosine/pharmacology
- Vascular Endothelial Growth Factor A/pharmacology
Collapse
Affiliation(s)
- Stéphanie Langlois
- Laboratoire de Médecine Moléculaire Ste-Justine-Université du Québec à Montréal, Centre de Cancérologie Charles-Bruneau, Hôpital Ste-Justine et Université du Québec à Montréal, Montréal, QC, Canada
| | | | | |
Collapse
|
823
|
Baudhuin LM, Jiang Y, Zaslavsky A, Ishii I, Chun J, Xu Y. S1P
3
‐mediated Akt activation and crosstalk with platelet‐derived growth factor receptor (PDGFR). FASEB J 2003; 18:341-3. [PMID: 14657000 DOI: 10.1096/fj.03-0302fje] [Citation(s) in RCA: 64] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Akt plays a pivotal role in cell survival and tumorigenesis. We investigated the potential interaction between sphingosine-1-phosphate (S1P) and platelet-derived growth factor (PDGF) in the Akt signaling pathway. Using mouse embryonic fibroblasts (MEFs) from S1P receptor knockout mice, we show here that S1P3 was required for S473 phosphorylation of Akt by S1P. In addition, S1P-stimulated activation of Akt, but not ERK, was blocked by a PDGF receptor (PDGFR)-specific inhibitor, AG1296, suggesting a S1P3-mediated specific crosstalk between the Akt signaling pathways of S1P and PDGFR in MEFs. We investigated this crosstalk under different conditions and found that both Akt and ERK activation induced by S1P, but not lysophosphatidic acid (LPA), in HEY ovarian cancer cells required PDGFR but not epidermal growth factor receptor (EGFR) or insulin-like growth factor-I receptor (IGFR). Importantly, S1P induced a Gi-dependent tyrosine phosphorylation of PDGFR in HEY cells. This dependence on PDGFR in S1P-induced Akt activation was also observed in A2780, T47D, and HMEC-1 cells (which express S1P3), but not in PC-3 or GI-101A cells (which do not express S1P3), further supporting that S1P3 mediates the crosstalk between S1P and PDGFR. This is the first report demonstrating a unique interaction between S1P3 and PDGFR, in addition to demonstrating a specific role for S1P3 in S1P-induced Akt activation.
Collapse
Affiliation(s)
- Linnea M Baudhuin
- Department of Cancer Biology, The Lerner Research Institute, The Cleveland Clinic Foundation, Cleveland, Ohio 44195, USA
| | | | | | | | | | | |
Collapse
|
824
|
Higgins JPT, Wang L, Kambham N, Montgomery K, Mason V, Vogelmann SU, Lemley KV, Brown PO, Brooks JD, van de Rijn M. Gene expression in the normal adult human kidney assessed by complementary DNA microarray. Mol Biol Cell 2003; 15:649-56. [PMID: 14657249 PMCID: PMC329285 DOI: 10.1091/mbc.e03-06-0432] [Citation(s) in RCA: 84] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
The kidney is a highly specialized organ with a complex, stereotyped architecture and a great diversity of functions and cell types. Because the microscopic organization of the nephron, the functional unit of the kidney, has a consistent relationship to the macroscopic anatomy of the kidney, knowledge of the characteristic patterns of gene expression in different compartments of the kidney could provide insight into the functions and functional organization of the normal nephron. We studied gene expression in dissected renal lobes of five adult human kidneys using cDNA microarrays representing approximately 30,000 different human genes. Total RNA was isolated from sections of the inner and outer cortex, inner and outer medulla, papillary tips, and renal pelvis and from glomeruli isolated by sieving. The results revealed unique and highly distinctive patterns of gene expression for glomeruli, cortex, medulla, papillary tips, and pelvic samples. Immunohistochemical staining using selected antisera confirmed differential expression of several cognate proteins and provided histological localization of expression within the nephron. The distinctive patterns of gene expression in discrete portions of the kidney may serve as a resource for further understanding of renal physiology and the molecular and cellular organization of the nephron.
Collapse
Affiliation(s)
- John P T Higgins
- Department of Pathology, Stanford University School of Medicine, Stanford, California 94305, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
825
|
Abstract
Successful sequencing of the human genome has opened a new era in the life sciences and has greatly accelerated biomedical research. Among various research endeavors benefiting from established genomic information, one of the most fruitful areas is the research on orphan G protein-coupled receptors (GPCRs). Many intercellular mediators, including peptides, lipids, and other small molecules, have found their GPCRs in the plasma membrane, e.g., relaxin and tyramine. In the past 14 months, more than one dozen papers have been published reporting the finding of intercellular lipid mediators acting on rhodopsin family GPCRs. This review focuses primarily on intercellular lipid mediators and their recently discovered GPCRs.
Collapse
Affiliation(s)
- Dong-Soon Im
- Laboratory of Pharmacology, College of Pharmacy, Pusan National University, San 30, Chang-Jun-dong, Keum-Jung-gu, Busan 609-735, Republic of Korea.
| |
Collapse
|
826
|
Sanchez T, Estrada-Hernandez T, Paik JH, Wu MT, Venkataraman K, Brinkmann V, Claffey K, Hla T. Phosphorylation and action of the immunomodulator FTY720 inhibits vascular endothelial cell growth factor-induced vascular permeability. J Biol Chem 2003; 278:47281-90. [PMID: 12954648 DOI: 10.1074/jbc.m306896200] [Citation(s) in RCA: 317] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
FTY720, a potent immunosuppressive agent, is phosphorylated in vivo into FTY720-P, a high affinity agonist for sphingosine 1-phosphate (S1P) receptors. The effects of FTY720 on vascular cells, a major target of S1P action, have not been addressed. We now report the metabolic activation of FTY720 by sphingosine kinase-2 and potent activation of vascular endothelial cell functions in vitro and in vivo by phosphorylated FTY720 (FTY720-P). Incubation of endothelial cells with FTY720 resulted in phosphorylation by sphingosine kinase activity and formation of FTY720-P. Sphingosine kinase-2 effectively phosphorylated FTY720 in the human embryonic kidney 293T heterologous expression system. FTY720-P treatment of endothelial cells stimulated extracellular signal-activated kinase and Akt phosphorylation and adherens junction assembly and promoted cell survival. The effects of FTY720-P were inhibited by pertussis toxin, suggesting the requirement for Gi-coupled S1P receptors. Indeed, transmonolayer permeability induced by vascular endothelial cell growth factor was potently reversed by FTY720-P. Furthermore, oral FTY720 administration in mice potently blocked VEGF-induced vascular permeability in vivo. These findings suggest that FTY720 or its analogs may find utility in the therapeutic regulation of vascular permeability, an important process in angiogenesis, inflammation, and pathological conditions such as sepsis, hypoxia, and solid tumor growth.
Collapse
Affiliation(s)
- Teresa Sanchez
- Center for Vascular Biology, Department of Cell Biology, University of Connecticut Health Center, 263 Farmington Avenue, Farmington, CT 06030-3501, USA
| | | | | | | | | | | | | | | |
Collapse
|
827
|
Olivera A, Rosenfeldt HM, Bektas M, Wang F, Ishii I, Chun J, Milstien S, Spiegel S. 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; 278:46452-60. [PMID: 12963721 DOI: 10.1074/jbc.m308749200] [Citation(s) in RCA: 128] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Sphingosine 1-phosphate (S1P) is the ligand for a family of specific G protein-coupled receptors (GPCRs) that regulate a wide variety of important cellular functions, including growth, survival, cytoskeletal rearrangements, and cell motility. However, whether it also has an intracellular function is still a matter of great debate. Overexpression of sphingosine kinase type 1, which generated S1P, induced extensive stress fibers and impaired formation of the Src-focal adhesion kinase signaling complex, with consequent aberrant focal adhesion turnover, leading to inhibition of cell locomotion. We have dissected biological responses dependent on intracellular S1P from those that are receptor-mediated by specifically blocking signaling of Galphaq, Galphai, Galpha12/13, and Gbetagamma subunits, the G proteins that S1P receptors (S1PRs) couple to and signal through. We found that intracellular S1P signaled "inside out" through its cell-surface receptors linked to G12/13-mediated stress fiber formation, important for cell motility. Remarkably, cell growth stimulation and suppression of apoptosis by endogenous S1P were independent of GPCRs and inside-out signaling. Using fibroblasts from embryonic mice devoid of functional S1PRs, we also demonstrated that, in contrast to exogenous S1P, intracellular S1P formed by overexpression of sphingosine kinase type 1 promoted growth and survival independent of its GPCRs. Hence, exogenous and intracellularly generated S1Ps affect cell growth and survival by divergent pathways. Our results demonstrate a receptor-independent intracellular function of S1P, reminiscent of its action in yeast cells that lack S1PRs.
Collapse
Affiliation(s)
- Ana Olivera
- Molecular Immunology and Inflammation Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases/NIH, Bethesda, MD 20891, USA
| | | | | | | | | | | | | | | |
Collapse
|
828
|
Allende ML, Yamashita T, Proia RL. G-protein-coupled receptor S1P1 acts within endothelial cells to regulate vascular maturation. Blood 2003; 102:3665-7. [PMID: 12869509 DOI: 10.1182/blood-2003-02-0460] [Citation(s) in RCA: 295] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Sphingosine-1-phosphate (S1P) stimulates signaling pathways via G-protein-coupled receptors and triggers diverse cellular processes, including growth, survival, and migration. In S1P1 receptor-deficient embryos, blood vessels were incompletely covered by vascular smooth muscle cells (VSMCs), indicating the S1P1 receptor regulates vascular maturation. Because S1P1 receptor expression is not restricted to a particular cell type, it was not known whether the S1P1 receptor controlled VSMC coverage of vessels in a cell-autonomous fashion by functioning directly in VSMCs or indirectly through its activity in endothelial cells (ECs). By using the Cre/loxP system, we disrupted the S1P1 gene solely in ECs. The phenotype of the conditional mutant embryos mimicked the one obtained in the embryos globally deficient in S1P1. Thus, vessel coverage by VSMCs is directed by the activity of the S1P1 receptor in ECs.
Collapse
Affiliation(s)
- Maria L Allende
- Genetics of Development and Disease Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | | | | |
Collapse
|
829
|
Jarvis JN, Dozmorov I, Jiang K, Frank MB, Szodoray P, Alex P, Centola M. Novel approaches to gene expression analysis of active polyarticular juvenile rheumatoid arthritis. Arthritis Res Ther 2003; 6:R15-R32. [PMID: 14979934 PMCID: PMC400410 DOI: 10.1186/ar1018] [Citation(s) in RCA: 64] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2003] [Revised: 09/05/2003] [Accepted: 10/02/2003] [Indexed: 12/12/2022] Open
Abstract
Juvenile rheumatoid arthritis (JRA) has a complex, poorly characterized pathophysiology. Modeling of transcriptosome behavior in pathologic specimens using microarrays allows molecular dissection of complex autoimmune diseases. However, conventional analyses rely on identifying statistically significant differences in gene expression distributions between patients and controls. Since the principal aspects of disease pathophysiology vary significantly among patients, these analyses are biased. Genes with highly variable expression, those most likely to regulate and affect pathologic processes, are excluded from selection, as their distribution among healthy and affected individuals may overlap significantly. Here we describe a novel method for analyzing microarray data that assesses statistically significant changes in gene behavior at the population level. This method was applied to expression profiles of peripheral blood leukocytes from a group of children with polyarticular JRA and healthy control subjects. Results from this method are compared with those from a conventional analysis of differential gene expression and shown to identify discrete subsets of functionally related genes relevant to disease pathophysiology. These results reveal the complex action of the innate and adaptive immune responses in patients and specifically underscore the role of IFN-γ in disease pathophysiology. Discriminant function analysis of data from a cohort of patients treated with conventional therapy identified additional subsets of functionally related genes; the results may predict treatment outcomes. While data from only 9 patients and 12 healthy controls was used, this preliminary investigation of the inflammatory genomics of JRA illustrates the significant potential of utilizing complementary sets of bioinformatics tools to maximize the clinical relevance of microarray data from patients with autoimmune disease, even in small cohorts.
Collapse
Affiliation(s)
- James N Jarvis
- Department of Pediatrics, University of Oklahoma College of Medicine, Oklahoma City, OK, USA.
| | | | | | | | | | | | | |
Collapse
|
830
|
Favre CJ, Mancuso M, Maas K, McLean JW, Baluk P, McDonald DM. Expression of genes involved in vascular development and angiogenesis in endothelial cells of adult lung. Am J Physiol Heart Circ Physiol 2003; 285:H1917-38. [PMID: 12842817 DOI: 10.1152/ajpheart.00983.2002] [Citation(s) in RCA: 108] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Profiling gene expression in endothelial cells advances the understanding of normal vascular physiology and disease processes involving angiogenesis. However, endothelial cell purification has been challenging because of the difficulty of isolating cells and their low abundance. Here we examine gene expression in endothelial cells freshly isolated from lung capillaries after in vivo labeling with fluorescent cationic liposomes and purification by fluorescence-activated cell sorting (FACS). Of the 39,000 genes and expressed sequence tags evaluated on custom oligonucleotide arrays, 555 were enriched in endothelial cell fraction. These included familiar endothelial cell-associated genes such as VEGF, VEGF receptor (VEGFR)-1, VEGFR-2, angiopoietin-2, Tie1, Tie2, Edg1 receptor, VE-cadherin, claudin 5, connexin37, CD31, and CD34. Also enriched were genes in semaphorin/neuropilin (Sema3c and Nrp1), ephrin/Eph (ephrin A1, B1, B2, and EphB4), delta/notch (Hey1, Jagged 2, Notch 1, Notch 4, Numb, and Siah1b), and Wingless (Frizzled-4 and Tle1) signaling pathways involved in vascular development and angiogenesis. Expression of representative genes in alveolar capillary endothelial cells was verified by immunohistochemistry. Such expression reflects features that endothelial cells of normal lung capillaries have in common with embryonic and growing blood vessels. About half of the enriched genes, including exostosin 2, lipocalin 7, phospholipid scramblase 2, pleckstrin 2, protocadherin 1, Ryk, scube 1, serpinh1, SNF-related kinase, and several tetraspanins, had little or no previous association with endothelial cells. This approach can readily be used to profile genes expressed in blood vessels in tumors, chronic inflammation, and other sites in which endothelial cells avidly take up cationic liposomes.
Collapse
Affiliation(s)
- Cecile J Favre
- Department of Anatomy S1363, University of California-San Francisco, 513 Parnassus Avenue, San Francisco, CA 94143-0452, USA
| | | | | | | | | | | |
Collapse
|
831
|
Coussin F, Scott RH, Nixon GF. Sphingosine 1-phosphate induces CREB activation in rat cerebral artery via a protein kinase C-mediated inhibition of voltage-gated K+ channels. Biochem Pharmacol 2003; 66:1861-70. [PMID: 14563496 DOI: 10.1016/s0006-2952(03)00546-x] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Sphingosine 1-phosphate (S1P) is a potential mitogenic stimulus for vascular smooth muscle. S1P promotes an increase in the intracellular calcium concentration ([Ca(2+)](i)) in cerebral arteries, however S1P effects on regulation of gene expression are not known. Activation of the Ca(2+)-dependent transcription factor, cAMP response element-binding protein (CREB), is associated with smooth muscle proliferation. The aim of this study was to examine the Ca(2+)-dependent mechanisms involved in S1P-induced CREB activation in cerebral artery. Western blotting and immunofluorescence with a phospho-CREB antibody were used to detect CREB activation in Sprague-Dawley rat cerebral arteries. Whole-cell patch clamp recording and single cell imaging of [Ca(2+)](i) were performed on freshly isolated cerebral artery myocytes. S1P increased activation of CREB in the nucleus of cerebral arteries. This activation was mediated by Ca(2+)/calmodulin-dependent protein kinase and was dependent on an increase in [Ca(2+)](i) via two mechanisms: (i) intracellular Ca(2+) release via an inositol 1,4,5-trisphosphate (InsP(3))-dependent pathway and (ii) Ca(2+) entry through voltage-dependent Ca(2+) channels (VDCC). Activation of the VDCC occurred through S1P-induced inhibition (approximately 50%) of the voltage-gated potassium (K(+)) current. This inhibition was via a protein kinase C-mediated pathway resulting in tyrosine phosphorylation of at least one isoform of the Kv channel (Kv 1.2). These results demonstrate that S1P can activate the transcription factor CREB through different Ca(2+)-dependent pathways including intracellular Ca(2+) release and inhibition of voltage-gated K(+) channels leading to Ca(2+) influx. Our findings suggest a potential role for S1P in regulation of gene expression in vascular smooth muscle.
Collapse
Affiliation(s)
- Frederic Coussin
- Department of Biomedical Sciences, Institute of Medical Sciences, University of Aberdeen, Foresterhill, Aberdeen AB25 2ZD, UK
| | | | | |
Collapse
|
832
|
Liu H, Toman RE, Goparaju SK, Maceyka M, Nava VE, Sankala H, Payne SG, Bektas M, Ishii I, Chun J, Milstien S, Spiegel S. Sphingosine kinase type 2 is a putative BH3-only protein that induces apoptosis. J Biol Chem 2003; 278:40330-6. [PMID: 12835323 DOI: 10.1074/jbc.m304455200] [Citation(s) in RCA: 293] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
There are two isoforms of sphingosine kinase (SphK) that catalyze the formation of sphingosine 1-phosphate, a potent sphingolipid mediator. Whereas SphK1 stimulates growth and survival, here we show that SphK2 enhanced apoptosis in diverse cell types and also suppressed cellular proliferation. Apoptosis was preceded by cytochrome c release and activation of caspase-3. SphK2-induced apoptosis was independent of activation of sphingosine 1-phosphate receptors. Sequence analysis revealed that SphK2 contains a 9-amino acid motif similar to that present in BH3-only proteins, a pro-apoptotic subgroup of the Bcl-2 family. As with other BH3-only proteins, co-immunoprecipitation demonstrated that SphK2 interacted with Bcl-xL. Moreover, site-directed mutation of Leu-219, the conserved leucine residue present in all BH3 domains, markedly suppressed SphK2-induced apoptosis. Hence, the apoptotic effect of SphK2 might be because of its putative BH3 domain.
Collapse
Affiliation(s)
- Hong Liu
- Department of Biochemistry, Medical College of Virginia Campus, Virginia Commonwealth University, Richmond, Virginia 23298-0614, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
833
|
Abstract
Blood lymphocyte numbers, which are maintained by recirculation through secondary lymphoid organs, are essential for the efficient development of immune responses. Recirculating populations of B and T lymphocytes are regulated by the sphingosine-1-phosphate (S1P) receptor-dependent control of lymphocyte egress. T-cell egress from thymus into blood, egress from lymph node and Peyer's patch into lymph, and B-cell egress into lymph are rapidly and completely inhibited by agonism of S1P receptors. Mesenteric lymph nodes show log-jamming of lymphocytes subjacent to sinus-lining endothelium. Agonism of S1P receptors produces rapid peripheral blood lymphopenia, which is maintained in the presence of receptor agonist. Effector CD4+ and CD8+ T cells, produced by clonal expansion in draining lymph node in response to antigen, are sequestered in lymph node and fail to reach the peripheral blood. The S1P receptor system may represent an early physiological link between the non-specific inflammatory response and the alteration of lymphocyte traffic through draining lymph nodes. Pharmacological subversion of the S1P receptor system, through systemic S1P agonist-induced inhibition of lymphocyte egress, suppresses antigenic responses to peripheral, but not to systemically, delivered antigen. This inhibition induces significant immunosuppression in models of transplantation and autoimmune tissue damage that may prove to be of clinical benefit.
Collapse
Affiliation(s)
- Hugh Rosen
- Department of Immunology, The Scripps Research Institute, La Jolla, CA 92037, USA.
| | | | | |
Collapse
|
834
|
Escalante-Alcalde D, Hernandez L, Le Stunff H, Maeda R, Lee HS, Sciorra VA, Daar I, Spiegel S, Morris AJ, Stewart CL. The lipid phosphatase LPP3 regulates extra-embryonic vasculogenesis and axis patterning. Development 2003; 130:4623-37. [PMID: 12925589 DOI: 10.1242/dev.00635] [Citation(s) in RCA: 140] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Bioactive phospholipids, which include sphingosine-1-phosphate, lysophosphatidic acid, ceramide and their derivatives regulate a wide variety of cellular functions in culture such as proliferation, apoptosis and differentiation. The availability of these lipids and their products is regulated by the lipid phosphate phosphatases (LPPs). Here we show that mouse embryos deficient for LPP3 fail to form a chorio-allantoic placenta and yolk sac vasculature. A subset of embryos also show a shortening of the anterior-posterior axis and frequent duplication of axial structures that are strikingly similar to the phenotypes associated with axin deficiency, a critical regulator of Wnt signaling. Loss of LPP3 results in a marked increase in beta-catenin-mediated TCF transcription, whereas elevated levels of LPP3 inhibit beta-catenin-mediated TCF transcription. LPP3 also inhibits axis duplication and leads to mild ventralization in Xenopus embryo development. Although LPP3 null fibroblasts show altered levels of bioactive phospholipids, consistent with loss of LPP3 phosphatase activity, mutant forms of LPP3, specifically lacking phosphatase activity, were able to inhibit beta-catenin-mediated TCF transcription and also suppress axis duplication, although not as effectively as intact LPP3. These results reveal that LPP3 is essential to formation of the chorio-allantoic placenta and extra-embryonic vasculature. LPP3 also mediates gastrulation and axis formation, probably by influencing the canonical Wnt signaling pathway. The exact biochemical roles of LPP3 phosphatase activity and its undefined effect on beta-catenin-mediated TCF transcription remain to be determined.
Collapse
Affiliation(s)
- Diana Escalante-Alcalde
- Cancer and Developmental Biology Laboratory, Division of Basic Science, National Cancer Institute, Frederick, MD 21702, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
835
|
Rosen H, Alfonso C, Surh CD, McHeyzer-Williams MG. Rapid induction of medullary thymocyte phenotypic maturation and egress inhibition by nanomolar sphingosine 1-phosphate receptor agonist. Proc Natl Acad Sci U S A 2003; 100:10907-12. [PMID: 12954982 PMCID: PMC196901 DOI: 10.1073/pnas.1832725100] [Citation(s) in RCA: 99] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2003] [Indexed: 11/18/2022] Open
Abstract
Only a small number of T cells generated in the thymus each day are selected to replenish the peripheral T cell pool. Much is known about thymic selection; however, little is known of the mechanisms regulating medullary maturation and the release of mature T cells into the blood. Here we demonstrate a rapid acceleration of medullary thymocyte phenotypic maturation through loss of CD69 induced by sphingosine 1-phosphate (S1P) receptor agonist. Low nanomolar agonist concentrations selectively induce changes in CD69(int) CD62L(high) single positive T cells, resulting in down-modulation of CD69 within 2 h. While CD69 loss is accelerated, egress of mature T cells into blood is inhibited >95% within 2 h. Both processes exhibit parallel sensitivities and dose-responses. Together, these data reveal a potent means for rapidly regulating thymic export where S1P receptor agonism alters both phenotypic maturation and egress of thymocytes into blood during late thymic maturation. The S1P system is now shown to acutely regulate both thymic and lymph node egress. Inhibition of lymphocyte egress from thymus and lymph node can contribute synergistically to clinically useful immunosupression by disrupting recirculation of peripheral T cells.
Collapse
Affiliation(s)
- Hugh Rosen
- Department of Immunology, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA 92037, USA.
| | | | | | | |
Collapse
|
836
|
Licht T, Tsirulnikov L, Reuveni H, Yarnitzky T, Ben-Sasson SA. Induction of pro-angiogenic signaling by a synthetic peptide derived from the second intracellular loop of S1P3 (EDG3). Blood 2003; 102:2099-107. [PMID: 12763936 DOI: 10.1182/blood-2002-12-3634] [Citation(s) in RCA: 83] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The G-protein-coupled receptors of the endothelial differentiation gene (EDG) family mediate pro-angiogenic activities, such as endothelial cell proliferation, chemotaxis, and vessel morphogenesis. We synthesized and tested the effects of a 9-amino acid peptide (KRX-725), derived from the second intracellular loop of S1P3 (EDG3). KRX-725 mimics the effects of sphingosine 1-phosphate (S1P), the natural ligand of S1P3, by triggering a Gi-dependent MEK-ERK (mitogen-activated protein kinase kinase and extracellular signal-regulated kinase) signal transduction pathway. Using aortic rings as an ex vivo model of angiogenesis, vascular sprouting was assessed in the presence of KRX-725 or S1P. KRX-725 induced extensive and dense vascular sprouts, which contain an elaborated organization of endothelial and smooth muscle layers, including lumen formation. When KRX-725 or S1P was combined with proangiogenic factors, such as basic fibroblast growth factor (bFGF), stem cell factor, or vascular endothelial growth factor, the effect was synergistic, leading to further enhancement of vascular sprouting. KRX-725 also initiated neovascularization in a mouse corneal pocket assay in vivo and showed synergism with bFGF. The specificity of KRX-725 was demonstrated via peptide-induced receptor internalization of S1P3 but not S1P1. The ability of a short peptide to stimulate extensive angiogenesis and to synergize with pro-angiogenic factors suggests that KRX-725 may serve as a useful agent in treating pathologic conditions such as peripheral vascular disease, cardiac ischemia, or tissue grafts.
Collapse
MESH Headings
- Animals
- Aorta/cytology
- Cells, Cultured
- Dose-Response Relationship, Drug
- Drug Synergism
- Endothelial Growth Factors/pharmacology
- Endothelium, Vascular/cytology
- Fibroblast Growth Factor 2/pharmacology
- GTP-Binding Protein alpha Subunits, Gi-Go/metabolism
- Humans
- I-kappa B Proteins/chemistry
- I-kappa B Proteins/genetics
- I-kappa B Proteins/metabolism
- Intercellular Signaling Peptides and Proteins/pharmacology
- Lymphokines/pharmacology
- MAP Kinase Signaling System/drug effects
- MAP Kinase Signaling System/physiology
- Mice
- Mice, Inbred C57BL
- Mitogen-Activated Protein Kinases/metabolism
- Muscle, Smooth, Vascular/cytology
- NF-KappaB Inhibitor alpha
- Neovascularization, Physiologic/drug effects
- Neovascularization, Physiologic/physiology
- Peptide Fragments/chemical synthesis
- Peptide Fragments/pharmacology
- Protein Structure, Tertiary
- Receptors, Lysophospholipid
- Stem Cell Factor/pharmacology
- Transfection
- Umbilical Veins/cytology
- Vascular Endothelial Growth Factor A
- Vascular Endothelial Growth Factors
Collapse
|
837
|
Fukuda Y, Kihara A, Igarashi Y. Distribution of sphingosine kinase activity in mouse tissues: contribution of SPHK1. Biochem Biophys Res Commun 2003; 309:155-60. [PMID: 12943676 DOI: 10.1016/s0006-291x(03)01551-1] [Citation(s) in RCA: 98] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Sphingosine kinase (SPHK) phosphorylates sphingosine to form a bioactive lipid mediator, sphingosine 1-phosphate (S1P). S1P mediates such diverse biological processes as regulation of cell differentiation, motility, and apoptosis both extracellularly, via S1P (Edg) family receptors, and intracellularly, through unidentified targets. In cells S1P is short-lived, so the synthetic process catalyzed by sphingosine kinase may be important in maintaining the cellular levels of the compound. Thus far, two sphingosine kinases have been reported, with SPHK1 exhibiting the higher activity. However, several studies suggest the existence of unidentified sphingosine kinases. Therefore, to further elucidate the role of SPHK1 in the formation of S1P, we investigated its contribution to the total sphingosine kinase activity in mouse tissues. We found that SPHK1 is a major sphingosine kinase in many tissues, especially in brain, heart, and colon. However, some tissues such as spleen, small intestine, and lung contained sphingosine kinase activity that was not attributable to SPHK1 or SPHK2, as determined by immunodepletion assays. Furthermore, the presence of other sphingosine kinases with different properties, i.e., higher activity toward phytosphingosine and a different subcellular distribution, is suggested.
Collapse
Affiliation(s)
- Yu Fukuda
- Department of Biomembrane and Biofunctional Chemistry, Graduate School of Pharmaceutical Sciences, Hokkaido University, Sapporo, Japan
| | | | | |
Collapse
|
838
|
Sato TN, Loughna S, Davis EC, Visconti RP, Richardson CD. Selective functions of angiopoietins and vascular endothelial growth factor on blood vessels: the concept of "vascular domain". COLD SPRING HARBOR SYMPOSIA ON QUANTITATIVE BIOLOGY 2003; 67:171-80. [PMID: 12858538 DOI: 10.1101/sqb.2002.67.171] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- T N Sato
- Sato Laboratory, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA
| | | | | | | | | |
Collapse
|
839
|
Arikawa K, Takuwa N, Yamaguchi H, Sugimoto N, Kitayama J, Nagawa H, Takehara K, Takuwa Y. Ligand-dependent inhibition of B16 melanoma cell migration and invasion via endogenous S1P2 G protein-coupled receptor. Requirement of inhibition of cellular RAC activity. J Biol Chem 2003; 278:32841-51. [PMID: 12810709 DOI: 10.1074/jbc.m305024200] [Citation(s) in RCA: 129] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
We investigated mechanisms for inhibition of B16 melanoma cell migration and invasion by sphingosine-1-phosphate (S1P), which is the ligand for the Edg family G protein-coupled receptors and also implicated as an intracellular second messenger. S1P, dihydro-S1P, and sphingosylphosphorylcholine inhibited B16 cell migration and invasion with the relative potencies expected as S1P2 receptor agonists. The S1P2-selective antagonist JTE013 completely abolished the responses to these agonists. In addition, JTE013 abrogated the inhibition by sphingosine, which is the S1P precursor but not an agonist for S1P receptors, indicating that the sphingosine effects were mediated via S1P2 stimulation, most likely by S1P that was converted from sphingosine. S1P induced inhibition and activation, respectively, of Rac and RhoA in B16 cells, which were abrogated by JTE013. Adenovirus-mediated expression of N17Rac mimicked S1P inhibition of migration, whereas C3 toxin pretreatment, but not Rho kinase inhibitors, reversed the S1P inhibition. Overexpression of S1P2 sensitized, and that of either S1P1 or S1P3 desensitized, B16 cells to S1P inhibition of Rac and migration. In JTE013-pretreated, S1P3-overexpressing B16 cells, S1P stimulated cellular RhoA but failed to inhibit either Rac or migration, indicating that RhoA stimulation itself is not sufficient for inhibition of migration. These results provide compelling evidence that endogenously expressed S1P2 negatively regulates cell motility and invasion through ligand-dependent reciprocal regulation of cellular Rac and RhoA activities. In the presence of JTE013, S1P instead stimulated Rac and migration in B16 cells that overexpress either S1P1 or S1P3, unveiling counteractions between S1P2 and S1P1 or S1P3 chemotactic receptor.
Collapse
Affiliation(s)
- Kayo Arikawa
- Department of Physiology, Kanazawa University Graduate School of Medicine, Kanazawa, Ishikawa 920-8640, Japan
| | | | | | | | | | | | | | | |
Collapse
|
840
|
Kohno T, Matsuyuki H, Inagaki Y, Igarashi Y. Sphingosine 1-phosphate promotes cell migration through the activation of Cdc42 in Edg-6/S1P4-expressing cells. Genes Cells 2003; 8:685-97. [PMID: 12875654 DOI: 10.1046/j.1365-2443.2003.00667.x] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
BACKGROUND Sphingosine 1-phosphate (Sph-1-P) is a bioactive lipid mediator released from activated platelets, which regulates diverse signal transduction pathways via cell surface receptors. Recent studies have revealed that the seven-transmembrane-spanning receptors, Edg-1, Edg-3, Edg-5, Edg-6 and Edg-8 are specific Sph-1-P receptors. Northern blot analysis has demonstrated that Edg-6 is expressed in lymphocyte-containing tissues such as spleen and lung. Little is known about the molecular mechanisms of Edg-6 functions, probably because of the difficulties in expressing Edg-6 on the cell surface. RESULTS Here, our studies revealed that N-terminal FLAG-tagged Edg-6 or Edg-6-GFP fusion protein was expressed in the endoplasmic reticulum, but was not expressed on the cell surface. On the other hand, C-terminally tagged Edg-6 or both N-terminally and C-terminally tagged Edg-6 was able to localize to the cell surface. Using these cells, we found that Sph-1-P induced cell migration through cell surface-expressed Edg-6 in a pertussis toxin-sensitive manner. This motility was mediated through the activation of a member of the Rho family of small GTPases, Cdc42. CONCLUSION These results support a role for Sph-1-P signalling via Edg-6 in the pathways involved in cell motility.
Collapse
Affiliation(s)
- Takayuki Kohno
- Department of Biomembrane and Biofunctional Chemistry, Graduate School of Pharmaceutical Sciences, Hokkaido University, Nishi 6, Kita 12, Kita-ku, Sapporo 060-0812 Japan
| | | | | | | |
Collapse
|
841
|
Abstract
New insights have been gained into the therapeutic relevance of the sphingosine 1-phosphate (S1P) pathway, on the basis of reverse pharmacological approaches to defining the mechanism of action of the immunosuppressive agent FTY720. Natural and synthetic sphingosine 1-phosphate receptor agonists can make picomolar interactions with their cognate G-protein-coupled receptors, and provide chemical approaches to defining the contribution of distinct receptor subtypes to pathology, physiology and treatment. The chemistry of S1P receptors and their synthetic ligands present a paradigm for the understanding of lipid-receptor interactions and their contribution to physiology and pathology. These approaches can potentially be extended to a broad, related family of G-protein-coupled receptors that share ligands and ligand similarities.
Collapse
Affiliation(s)
- Hugh Rosen
- Department of Immunology and Committee for Advanced Human Therapeutics, The Scripps Research Institute, ICND-118 10550 North Torrey Pines Road, La Jolla, CA 92037, USA.
| | | |
Collapse
|
842
|
zu Heringdorf DM, Vincent MEM, Lipinski M, Danneberg K, Stropp U, Wang DA, Tigyi G, Jakobs KH. Inhibition of Ca(2+) signalling by the sphingosine 1-phosphate receptor S1P(1). Cell Signal 2003; 15:677-87. [PMID: 12742228 DOI: 10.1016/s0898-6568(03)00011-1] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The lysophospholipid, sphingosine 1-phosphate (S1P), regulates a multitude of cellular functions by activating specific G protein-coupled receptors (GPCRs) (S1P(1-5), plus three newly identified S1P receptors). The G(i)-coupled S1P(1) receptor inhibits adenylyl cyclase, stimulates mitogen-activated protein kinases (MAP kinases) and cell migration, and is required for blood vessel maturation. Here, we report that S1P(1) inhibits Ca(2+) signalling in a number of cell types. In HEK-293 cells, which endogenously express S1P(1-3), overexpression of S1P(1) reduced intracellular free Ca(2+) concentration ([Ca(2+)](i)) increases induced by various receptor agonists as well as thapsigargin. The inhibitory Ca(2+) signalling of S1P(1) was blocked by pertussis toxin (PTX) and the protein kinase C (PKC) inhibitor, Gö6976, and imitated by phorbol ester and overexpression of classical PKC isoforms. Activation of S1P(1) stably expressed in RH7777 cells, which endogenously do not express S1P receptors, also inhibited Ca(2+) signalling, without mediating Ca(2+) mobilization on its own. It is concluded that the widely expressed S1P receptor S1P(1) inhibits Ca(2+) signalling, most likely via G(i) proteins and classical PKC isoforms. Co-expression of S1P(1) with S1P(3), but not S1P(2), reversed the inhibitory effect of S1P(1), furthermore suggesting a specific interplay of S1P receptor subtypes usually found within a single cell type.
Collapse
|
843
|
Yopp AC, Randolph GJ, Bromberg JS. Leukotrienes, sphingolipids, and leukocyte trafficking. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2003; 171:5-10. [PMID: 12816975 DOI: 10.4049/jimmunol.171.1.5] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Affiliation(s)
- Adam C Yopp
- Carl C. Icahn Center for Gene Therapy and Molecular Medicine and Recanti/Miller Transplantation Institute, Mount Sinai School of Medicine, New York, NY 10029, USA
| | | | | |
Collapse
|
844
|
Annabi B, Thibeault S, Lee YT, Bousquet-Gagnon N, Eliopoulos N, Barrette S, Galipeau J, Béliveau R. Matrix metalloproteinase regulation of sphingosine-1-phosphate-induced angiogenic properties of bone marrow stromal cells. Exp Hematol 2003; 31:640-649. [PMID: 12842709 DOI: 10.1016/s0301-472x(03)00090-0] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/09/2023]
Abstract
OBJECTIVE Bone marrow-derived stromal cells (MSC) are able to acquire histological and immunophenotypic characteristics consistent with endothelial cells (EC). In this study we examined the effect of sphingosine-1-phosphate (S1P), a platelet-derived bioactive lysophospholipid that is believed to specifically stimulate EC migration and tube formation, on the angiogenic properties of MSC. METHODS MSC were isolated from murine bone marrow and cultured in the presence of diverse angiogenic growth factors. Using a chemotaxis chamber and Matrigel tubulogenesis assay, we measured the extent of MSC migration and capillary-like structure formation. Western blots and zymography were used to assess the levels and activation states of soluble and membrane-bound matrix metalloproteinase (MMP). RESULTS We found that S1P strongly induced MSC migration and in vitro capillary-like structure formation. Ilomastat, a broad-spectrum MMP inhibitor, antagonized several angiogenic and S1P-mediated events in MSC. These included 1) the inhibition of S1P-induced tube formation, 2) the inhibition of concanavalin-A (Con-A)-mediated proMMP-2 activation, and 3) the inhibition of S1P- and Con-A-induced caspase-3 activity. Moreover, S1P induced membrane type-1 (MT1)-MMP mRNA and protein expression, but paradoxically antagonized its cell surface proteolytic processing. In addition, anti-angiogenic agents such as Ilomastat, Neovastat, and green tea polyphenol epigallocatechin-3-gallate antagonized the S1P-induced migration of MSC as well as that of transfected COS-7 cells overexpressing the recombinant receptor for S1P, EDG-1. CONCLUSION Collectively, our results indicate a crucial role for S1P/EDG-1-mediated angiogenic and survival events in the regulation of microvascular network remodeling by MSC, and may provide a new molecular link between hemostasis and angiogenesis processes.
Collapse
Affiliation(s)
- Borhane Annabi
- Laboratoire de Médecine Moléculaire, Centre de Cancérologie Charles-Bruneau, Hôpital Sainte-Justine and Université du Québec à Montréal, Montreal, Quebec, Canada
| | | | | | | | | | | | | | | |
Collapse
|
845
|
Abstract
Sphingosine-1-phosphate (SIP) is a bioactive sphingolipid metabolite that regulates diverse cellular responses including, growth, survival, cytoskeleton rearrangements and movement. SIP plays an important role during development, particularly in vascular maturation and has been implicated in pathophysiology of cancer, wound healing, and atherosclerosis. This review summarizes the evidence showing that signaling induced by SIP is complex and involves both intracellular and extracellular actions. The intracellular effects of SIP remain speculative awaiting the identification of specific targets whereas the extracellular effects of SIP are clearly mediated through the activation of five specific G protein coupled receptors, called S1P1-5. Recent studies demonstrate that intracellular generated SIP can act in a paracrine or autocrine manner to activate its cell surface receptors.
Collapse
Affiliation(s)
- Kenneth Watterson
- Department of Biochemistry, Medical College of Virginia Campus, Virginia Commonwealth University, Tichmond, VA 23298, USA
| | | | | | | |
Collapse
|
846
|
Fegley AJ, Tanski WJ, Roztocil E, Davies MG. Sphingosine-1-phosphate stimulates smooth muscle cell migration through galpha(i)- and pi3-kinase-dependent p38(MAPK) activation. J Surg Res 2003; 113:32-41. [PMID: 12943808 DOI: 10.1016/s0022-4804(03)00120-3] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
BACKGROUND Sphingosine-1-phosphate (S-1-P) is an extracellular mediator released in response to vessel injury. S-1-P binds to G-protein-coupled receptors, which can be Galpha(i)-, Galpha(q)-, or G(12/13)-linked. This study examines the role of p38 mitogen-activated protein kinase (p38(MAPK)) in vascular smooth muscle cell migration after stimulation with S-1-P, and pathways leading to p38(MAPK) activation. S-1-P has previously been shown to stimulate migration of vascular smooth muscle cells (VSMCs) in vitro through ERK1/2 and G(i). We hypothesized that S-1-P-induced VSMC migration is also dependent on p38(MAPK) activation through a G(i)-coupled extracellular receptor and phosphoinositide 3-kinase (PI3-K). METHODS VSMCs were cultured in vitro. A linear wound assay was performed in the presence of S-1-P and inhibitors of p38(MAPK) (SB203580) or epidermal growth factor (EGF) receptor kinase (AG1478). Chemotaxis stimulated by S-1-P was also assayed in a modified Boyden chamber with and without SB203580 pretreatment. Western blotting was performed to examine p38(MAPK) activation in response to S-1-P with and without SB203580, AG1478, or inhibitors of G(i) (pertussis toxin), PI3-K (Wortmannin and LY294002), or MEK1 (PD98059). Western blotting and immunoprecipitation for targets of p38(MAPK) (MAPKAP kinase-2) and PI3-K (Akt) were also performed.S-1-P stimulated migration of VSMCs in both wound and Boyden transwell assays. This migration was inhibited by SB203580 to the level of control, whereas AG478 had no effect. RESULTS S-1-P stimulated activation of p38(MAPK) that peaked at 10 min, as well as activation of MAPKAP kinase-2. Activation of p38(MAPK) was significantly inhibited by SB203580, pertussis toxin, Wortmannin, and LY294002, but not by PD98059 or AG1478; MAPKAP kinase-2 activation was inhibited by SB203580. Akt was activated by S-1-P at 3 to 5 min; this response was inhibited by Wortmannin and LY294002, but not by SB203580 or pertussis toxin. CONCLUSIONS S-1-P induced VSMC migration through a G(i)-linked and a PI3-K coupled, p38(MAPK)- dependent process. PI3-K appears to function upstream of p38(MAPK), but was not G(i)-dependent. S-1-P-stimulated activation of p38(MAPK) does not signal via transactivation of the EGF receptor. Understanding signal transduction will allow targeted molecular interventions to treat the response of a vessel to injury.
Collapse
Affiliation(s)
- Allison J Fegley
- Vascular Biology and Therapeutics Program, Division of Vascular Surgery, Department of Surgery, and Center for Cardiovascular Research, University of Rochester, Rochester, New York, USA
| | | | | | | |
Collapse
|
847
|
Schaphorst KL, Chiang E, Jacobs KN, Zaiman A, Natarajan V, Wigley F, Garcia JGN. Role of sphingosine-1 phosphate in the enhancement of endothelial barrier integrity by platelet-released products. Am J Physiol Lung Cell Mol Physiol 2003; 285:L258-67. [PMID: 12626332 DOI: 10.1152/ajplung.00311.2002] [Citation(s) in RCA: 133] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
In vitro and in vivo evidence indicates that circulating platelets affect both vascular integrity and hemostasis. How platelets enhance the permeability barrier of the vascular endothelium is not well understood. We measured the effect of isolated human platelets on human pulmonary artery endothelial cell (EC) barrier integrity by monitoring transmonolayer electrical resistance. EC barrier function was significantly increased by the addition of platelets ( approximately 40% maximum, 2.5 x 106 platelets/ml). Platelet supernatants, derived from 2.5 x 106 platelets/ml, reproduced the barrier enhancement and reversed the barrier dysfunction produced by the edemagenic agonist thrombin, which implicates a soluble barrier-promoting factor. The barrier-enhancing effect of platelet supernatants was heat stable but was attenuated by either charcoal delipidation (suggesting a vasoactive lipid mediator) or pertussis toxin, implying involvement of a Gialpha-coupled receptor signal transduction pathway. Sphingosine-1-phosphate (S1P), a sphingolipid that is released from activated platelets, is known to ligate G protein-coupled EC differentiation gene (EDG) receptors, increase EC electrical resistance, and reorganize the actin cytoskeleton (Garcia JG, Liu F, Verin AD, Birukova A, Dechert MA, Gerthoffer WT, Bamberg JR, and English D. J Clin Invest 108: 689-701, 2001). Infection of EC with an adenoviral vector expressing an antisense oligonucleotide directed against EDG-1 but not infection with control vector attenuated the barrier-enhancing effect of both platelet supernatants and S1P. These results indicate that a major physiologically relevant vascular barrier-protective mediator produced by human platelets is S1P.
Collapse
Affiliation(s)
- Kane L Schaphorst
- Division of Pulmonary and Critical Care Medicine and Center for Translational Respiratory Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland 21224-6801, USA.
| | | | | | | | | | | | | |
Collapse
|
848
|
Mendel J, Heinecke K, Fyrst H, Saba JD. Sphingosine phosphate lyase expression is essential for normal development in Caenorhabditis elegans. J Biol Chem 2003; 278:22341-9. [PMID: 12682045 DOI: 10.1074/jbc.m302857200] [Citation(s) in RCA: 57] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Sphingolipids are ubiquitous membrane constituents whose metabolites function as signaling molecules in eukaryotic cells. Sphingosine 1-phosphate, a key sphingolipid second messenger, regulates proliferation, motility, invasiveness, and programmed cell death. These effects of sphingosine 1-phosphate and similar phosphorylated sphingoid bases have been observed in organisms as diverse as yeast and humans. Intracellular levels of sphingosine 1-phosphate are tightly regulated by the actions of sphingosine kinase, which is responsible for its synthesis and sphingosine-1-phosphate phosphatase and sphingosine phosphate lyase, the two enzymes responsible for its catabolism. In this study, we describe the cloning of the Caenorhabditis elegans sphingosine phosphate lyase gene along with its functional expression in Saccharomyces cerevisiae. Promoter analysis indicates tissue-specific and developmental regulation of sphingosine phosphate lyase gene expression. Inhibition of C. elegans sphingosine phosphate lyase expression by RNA interference causes accumulation of phosphorylated and unphosphorylated long-chain bases and leads to poor feeding, delayed growth, reproductive abnormalities, and intestinal damage similar to the effects seen with exposure to Bacillus thuringiensis toxin. Our results show that sphingosine phosphate lyase is an essential gene in C. elegans and suggest that the sphingolipid degradative pathway plays a conserved role in regulating animal development.
Collapse
Affiliation(s)
- Jane Mendel
- Children's Hospital Oakland Research Institute, Oakland, California 94609-1673, USA
| | | | | | | |
Collapse
|
849
|
Abstract
To most people, concerns over the link between lipids and cardiovascular health most likely end with monitoring their daily consumption of dietary fats. However, it has become increasingly clear that, in addition to effects on adult cardiovascular physiology, lipids also play key roles in the formation of a functioning cardiovascular system. The lysophospholipids, lysophosphatidic acid (LPA) and sphingosine 1-phosphate (S1P), have come to the forefront as developmental and physiological regulators of the cardiovascular system. In this review, we discuss the function of the G protein-coupled receptors responsible for transducing LPA and S1P signals during development of the vertebrate cardiovascular system, focusing first on their role in angiogenesis and then on their function during embryonic development.
Collapse
Affiliation(s)
- Nick Osborne
- Department of Biochemistry and Biophysics, University of California, San Francisco, California 94143-0448, USA.
| | | |
Collapse
|
850
|
Gräler MH, Grosse R, Kusch A, Kremmer E, Gudermann T, Lipp M. The sphingosine 1-phosphate receptor S1P4 regulates cell shape and motility via coupling to Gi and G12/13. J Cell Biochem 2003; 89:507-19. [PMID: 12761884 DOI: 10.1002/jcb.10537] [Citation(s) in RCA: 98] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Sphingosine 1-phosphate (S1P) receptors represent a novel subfamily of G-protein-coupled receptors binding S1P specifically and with high affinity. Although their in vivo functions remain largely unknown, in vitro extracellular application of S1P induces distinct S1P receptor-dependent cellular responses including proliferation, differentiation, and migration. We have analyzed signaling pathways engaged by S1P(4), which is highly expressed in the lymphoid system. Here we show that S1P(4) couples directly to Galpha(i) and even more effectively to Galpha(12/13)-subunits of trimeric G-proteins, but not to Galpha(q) unlike other S1P receptors. Consequently, CHO-K1 cells ectopically expressing S1P(4) potently activate the small GTPase Rho and undergo cytoskeletal rearrangements, inducing peripheral stress fiber formation and cell rounding, upon S1P stimulation. Overexpression of S1P(4) in Jurkat T cells induces pertussis toxin-sensitive cell motility even in the absence of exogenously added S1P. In addition, S1P(4) is internalized upon binding of S1P. The capacity of S1P(4) to mediate cellular responses, such as motility and shape change through Galpha(i)- and Galpha(12/13)-coupled signaling pathways may be important for its in vivo function which is currently under investigation.
Collapse
Affiliation(s)
- Markus H Gräler
- Department of Tumor Genetics and Immunogenetics, Max-Delbrück-Centrum of Molecular Medicine, Robert-Rössle-Strasse 10, 13092 Berlin, Germany
| | | | | | | | | | | |
Collapse
|