Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
Add more filters










Publication year range
1.
J Exp Med ; 216(7): 1582-1598, 2019 07 01.
Article in English | MEDLINE | ID: mdl-31147448

ABSTRACT

Sphingosine 1-phosphate (S1P) and lysophosphatidic acid (LPA) activate G protein-coupled receptors (GPCRs) to regulate biological processes. Using a genome-wide CRISPR/dCas9-based GPCR signaling screen, LPAR1 was identified as an inducer of S1PR1/ß-arrestin coupling while suppressing Gαi signaling. S1pr1 and Lpar1-positive lymphatic endothelial cells (LECs) of lymph nodes exhibit constitutive S1PR1/ß-arrestin signaling, which was suppressed by LPAR1 antagonism. Pharmacological inhibition or genetic loss of function of Lpar1 reduced the frequency of punctate junctions at sinus-lining LECs. Ligand activation of transfected LPAR1 in endothelial cells remodeled junctions from continuous to punctate structures and increased transendothelial permeability. In addition, LPAR1 antagonism in mice increased lymph node retention of adoptively transferred lymphocytes. These data suggest that cross-talk between LPAR1 and S1PR1 promotes the porous junctional architecture of sinus-lining LECs, which enables efficient lymphocyte trafficking. Heterotypic inter-GPCR coupling may regulate complex cellular phenotypes in physiological milieu containing many GPCR ligands.


Subject(s)
Endothelial Cells/metabolism , Lymph Nodes/metabolism , Receptor Cross-Talk , Animals , CRISPR-Associated Protein 9 , CRISPR-Cas Systems , Fluorescent Antibody Technique , Gene Editing , HEK293 Cells , Human Umbilical Vein Endothelial Cells , Humans , Lysophospholipids/metabolism , Mice , Real-Time Polymerase Chain Reaction , Receptors, G-Protein-Coupled/metabolism , Receptors, Lysophosphatidic Acid/metabolism , Sphingosine/analogs & derivatives , Sphingosine/metabolism , Sphingosine-1-Phosphate Receptors/metabolism
2.
Pharmacol Ther ; 193: 91-98, 2019 01.
Article in English | MEDLINE | ID: mdl-30048709

ABSTRACT

While normal angiogenesis is critical for development and tissue growth, pathological angiogenesis is important for the growth and spread of cancers by supplying nutrients and oxygen as well as providing a conduit for distant metastasis. The interaction among extracellular matrix molecules, tumor cells, endothelial cells, fibroblasts, and immune cells is critical in pathological angiogenesis, in which various angiogenic growth factors, chemokines, and lipid mediators produced from these cells as well as hypoxic microenvironment promote angiogenesis by regulating expression and/or activity of various related genes. Sphingosine 1-phosphate and lysophosphatidic acid, bioactive lipid mediators which act via specific G protein-coupled receptors, play critical roles in angiogenesis. In addition, other lipid mediators including prostaglandin E2, lipoxin, and resolvins are produced in a stimulus-dependent manner and have pro- or anti-angiogenic effects, presumably through their specific GPCRs. Dysregulated lipid mediator signaling pathways are observed in the contxt of some tumors. This review will focus on LPA and S1P, two bioactive lipid mediators in their regulation of angiogenesis and cell migration that are critical for tumor growth and spread.


Subject(s)
Lysophospholipids/metabolism , Neoplasms/metabolism , Neoplasms/pathology , Neovascularization, Pathologic/metabolism , Sphingosine/analogs & derivatives , Animals , Cell Movement , Humans , Sphingosine/metabolism
3.
Sci Rep ; 8(1): 4969, 2018 03 21.
Article in English | MEDLINE | ID: mdl-29563527

ABSTRACT

Sphingosine 1-phosphate (S1P) is an intercellular signaling molecule present in blood. Erythrocytes have a central role in maintaining the S1P concentration in the blood stream. We previously demonstrated that S1P is exported from erythrocytes by a glyburide-sensitive S1P transporter. However, the gene encoding the S1P transporter in erythrocytes is unknown. In this study, we found that the mouse erythroid cell line, MEDEP-E14, has S1P export activity and exhibits properties that are consistent with those of erythrocytes. Using microarray analysis of MEDEP-E14 cells and its parental cell line, E14TG2a, we identified several candidate genes for S1P export activity. Of those genes, only one gene, Mfsd2b, showed S1P transport activity. The properties of S1P release by MFSD2B were similar to those in erythrocytes. Moreover, knockout of MFSD2B in MEDEP-E14 cells decreased S1P export from the cells. These results strongly suggest that MFSD2B is a novel S1P transporter in erythroid cells.


Subject(s)
Erythroid Cells/metabolism , Lysophospholipids/metabolism , Membrane Proteins/metabolism , Sphingosine/analogs & derivatives , Animals , CHO Cells , Cell Line , Cricetulus , Gene Knockout Techniques , Membrane Proteins/genetics , Mice , Microarray Analysis , Phosphotransferases (Alcohol Group Acceptor)/genetics , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sphingosine/metabolism
4.
Elife ; 62017 09 28.
Article in English | MEDLINE | ID: mdl-28956531

ABSTRACT

Sphingosine-1-phosphate (S1P) is generated through phosphorylation of sphingosine by sphingosine kinases (Sphk1 and Sphk2). We show that sphk2 maternal-zygotic mutant zebrafish embryos (sphk2MZ) display early developmental phenotypes, including a delay in epiboly, depleted S1P levels, elevated levels of sphingosine, and resistance to sphingosine toxicity. The sphk2MZ embryos also have strikingly increased levels of maternal transcripts encoding ceramide synthase 2b (Cers2b), and loss of Cers2b in sphk2MZ embryos phenocopies sphingosine toxicity. An upstream region of the cers2b promoter supports enhanced expression of a reporter gene in sphk2MZ embryos compared to wildtype embryos. Furthermore, ectopic expression of Cers2b protein itself reduces activity of the promoter, and this repression is relieved by exogenous sphingosine. Therefore, the sphk2MZ genome recognizes the lack of sphingosine kinase activity and up-regulates cers2b as a salvage pathway for sphingosine turnover. Cers2b can also function as a sphingolipid-responsive factor to mediate at least part of a feedback regulatory mechanism.


Subject(s)
Homeostasis , Oxidoreductases/metabolism , Sphingosine/metabolism , Zebrafish/embryology , Animals , Gene Expression Regulation
5.
Sci Signal ; 10(492)2017 Aug 15.
Article in English | MEDLINE | ID: mdl-28811382

ABSTRACT

Endothelial dysfunction, a hallmark of vascular disease, is restored by plasma high-density lipoprotein (HDL). However, a generalized increase in HDL abundance is not beneficial, suggesting that specific HDL species mediate protective effects. Apolipoprotein M-containing HDL (ApoM+HDL), which carries the bioactive lipid sphingosine 1-phosphate (S1P), promotes endothelial function by activating G protein-coupled S1P receptors. Moreover, HDL-bound S1P is limiting in several inflammatory, metabolic, and vascular diseases. We report the development of a soluble carrier for S1P, ApoM-Fc, which activated S1P receptors in a sustained manner and promoted endothelial function. In contrast, ApoM-Fc did not modulate circulating lymphocyte numbers, suggesting that it specifically activated endothelial S1P receptors. ApoM-Fc administration reduced blood pressure in hypertensive mice, attenuated myocardial damage after ischemia/reperfusion injury, and reduced brain infarct volume in the middle cerebral artery occlusion model of stroke. Our proof-of-concept study suggests that selective and sustained targeting of endothelial S1P receptors by ApoM-Fc could be a viable therapeutic strategy in vascular diseases.


Subject(s)
Endothelium, Vascular/drug effects , Hypertension/prevention & control , Lysophospholipids/pharmacology , Receptors, Lysosphingolipid/metabolism , Reperfusion Injury/prevention & control , Sphingosine/analogs & derivatives , Animals , Apolipoproteins M/metabolism , Endothelium, Vascular/metabolism , Human Umbilical Vein Endothelial Cells , Humans , Hypertension/metabolism , Hypertension/pathology , Lipoproteins, HDL/metabolism , Male , Mice , Mice, Knockout , Protein Binding , Receptors, Fc/metabolism , Reperfusion Injury/metabolism , Reperfusion Injury/pathology , Signal Transduction/drug effects , Sphingosine/pharmacology
6.
Int J Mol Sci ; 17(5)2016 May 13.
Article in English | MEDLINE | ID: mdl-27187373

ABSTRACT

The zebrafish (Danio rerio) is an ideal vertebrate model to investigate the developmental molecular mechanism of organogenesis and regeneration. Recent innovation in genome editing technologies, such as zinc finger nucleases (ZFNs), transcription activator-like effector nucleases (TALENs) and the clustered regularly interspaced short palindromic repeats (CRISPR)/CRISPR associated protein 9 (Cas9) system, have allowed researchers to generate diverse genomic modifications in whole animals and in cultured cells. The CRISPR/Cas9 and TALEN techniques frequently induce DNA double-strand breaks (DSBs) at the targeted gene, resulting in frameshift-mediated gene disruption. As a useful application of genome editing technology, several groups have recently reported efficient site-specific integration of exogenous genes into targeted genomic loci. In this review, we provide an overview of TALEN- and CRISPR/Cas9-mediated site-specific integration of exogenous genes in zebrafish.


Subject(s)
Gene Editing/methods , Gene Knock-In Techniques/methods , Zebrafish/genetics , Animals , CRISPR-Cas Systems , Recombination, Genetic
7.
Genes Cells ; 20(8): 647-58, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26094551

ABSTRACT

The lipid mediator sphingosine-1-phosphate (S1P) regulates various physiological and pathological phenomena such as angiogenesis and oncogenesis. Secreted S1P associates with the G-protein-coupled S1P receptors (S1PRs), leading to the activation of downstream signaling molecules. In mammals, five S1prs have been identified and the genetic disruption of a single S1pr1 gene causes vascular defects. In zebrafish, seven s1prs have been isolated. We found that individual s1prs showed unique expression patterns with some overlapping expression domains during early embryogenesis. We generated all s1pr single-mutant zebrafish by introducing premature stop codons in their coding regions using transcription activator-like effector nucleases and analyzed their phenotypes during early embryogenesis. Zygotic s1pr1, s1pr3a, s1pr3b, s1pr4, s1pr5a and s1pr5b mutants showed no developmental defects and grew into adults, whereas zygotic s1pr2 mutant showed embryonic lethality with a cardiac defect, showing quite distinct embryonic phenotypes for individual S1pr mutants between zebrafish and mouse. We further generated maternal-zygotic s1pr1, s1pr3a, s1pr3b, s1pr4, s1pr5a and s1pr5b mutants and found that these maternal-zygotic mutants also showed no obvious developmental defects, presumably suggesting the redundant functions of the S1P receptor-mediated signaling in zebrafish.


Subject(s)
Embryonic Development , Mutation , Receptors, Lysosphingolipid/genetics , Zebrafish/embryology , Animal Fins/physiology , Animals , Mice , Receptors, Lysosphingolipid/metabolism , Zebrafish/genetics , Zebrafish/physiology
8.
J Biol Chem ; 290(24): 14841-51, 2015 Jun 12.
Article in English | MEDLINE | ID: mdl-25907554

ABSTRACT

Sphingosine 1-phosphate (S1P) is synthesized from sphingosine by sphingosine kinases (SPHK1 and SPHK2) in invertebrates and vertebrates, whereas specific receptors for S1P (S1PRs) selectively appear in vertebrates, suggesting that S1P acquires novel functions in vertebrates. Because the developmental functions of SPHK1 and SPHK2 remain obscure in vertebrates, we generated sphk1 or sphk2 gene-disrupted zebrafish by introducing premature stop codons in their coding regions using transcription activator-like effector nucleases. Both zygotic sphk1 and sphk2 zebrafish mutants exhibited no obvious developmental defects and grew to adults. The maternal-zygotic sphk2 mutant (MZsphk2), but not the maternal-zygotic sphk1 mutant and maternal sphk2 mutant, had a defect in the cardiac progenitor migration and a concomitant decrease in S1P level, leading to a two-heart phenotype (cardia bifida). Cardia bifida in MZsphk2, which was rescued by injecting sphk2 mRNA, was a phenotype identical to that of zygotic mutants of the S1P transporter spns2 and S1P receptor s1pr2, indicating that the Sphk2-Spns2-S1pr2 axis regulates the cardiac progenitor migration in zebrafish. The contribution of maternally supplied lipid mediators during vertebrate organogenesis presents as a requirement for maternal-zygotic Sphk2.


Subject(s)
Heart/embryology , Isoenzymes/metabolism , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Zygote/enzymology , Amino Acid Sequence , Animals , Female , Isoenzymes/chemistry , Isoenzymes/genetics , Molecular Sequence Data , Mutation , Phosphotransferases (Alcohol Group Acceptor)/chemistry , Phosphotransferases (Alcohol Group Acceptor)/genetics , Pregnancy , Sequence Homology, Amino Acid , Zebrafish
9.
Sci Rep ; 5: 8841, 2015 Mar 05.
Article in English | MEDLINE | ID: mdl-25740433

ABSTRACT

The CRISPR/Cas9 system provides a powerful tool for genome editing in various model organisms, including zebrafish. The establishment of targeted gene-disrupted zebrafish (knockouts) is readily achieved by CRISPR/Cas9-mediated genome modification. Recently, exogenous DNA integration into the zebrafish genome via homology-independent DNA repair was reported, but this integration contained various mutations at the junctions of genomic and integrated DNA. Thus, precise genome modification into targeted genomic loci remains to be achieved. Here, we describe efficient, precise CRISPR/Cas9-mediated integration using a donor vector harbouring short homologous sequences (10-40 bp) flanking the genomic target locus. We succeeded in integrating with high efficiency an exogenous mCherry or eGFP gene into targeted genes (tyrosinase and krtt1c19e) in frame. We found the precise in-frame integration of exogenous DNA without backbone vector sequences when Cas9 cleavage sites were introduced at both sides of the left homology arm, the eGFP sequence and the right homology arm. Furthermore, we confirmed that this precise genome modification was heritable. This simple method enables precise targeted gene knock-in in zebrafish.


Subject(s)
CRISPR-Cas Systems , Gene Knock-In Techniques , Zebrafish/genetics , Amino Acid Sequence , Animals , Base Sequence , Gene Targeting/methods , Genes, Reporter , Genetic Loci , Genetic Vectors/genetics , Genome , Homologous Recombination , Molecular Sequence Data
10.
Sci Rep ; 4: 6545, 2014 Oct 08.
Article in English | MEDLINE | ID: mdl-25293390

ABSTRACT

The type II bacterial CRISPR/Cas9 system is rapidly becoming popular for genome-engineering due to its simplicity, flexibility, and high efficiency. Recently, targeted knock-in of a long DNA fragment via homology-independent DNA repair has been achieved in zebrafish using CRISPR/Cas9 system. This raised the possibility that knock-in transgenic zebrafish could be efficiently generated using CRISPR/Cas9. However, how widely this method can be applied for the targeting integration of foreign genes into endogenous genomic loci is unclear. Here, we report efficient generation of knock-in transgenic zebrafish that have cell-type specific Gal4 or reporter gene expression. A donor plasmid containing a heat-shock promoter was co-injected with a short guide RNA (sgRNA) targeted for genome digestion, a sgRNA targeted for donor plasmid digestion, and Cas9 mRNA. We have succeeded in establishing stable knock-in transgenic fish with several different constructs for 4 genetic loci at a frequency being exceeding 25%. Due to its simplicity, design flexibility, and high efficiency, we propose that CRISPR/Cas9-mediated knock-in will become a standard method for the generation transgenic zebrafish.


Subject(s)
CRISPR-Cas Systems , Gene Knock-In Techniques , Genetic Engineering , Organisms, Genetically Modified/genetics , Zebrafish/genetics , Animals , Animals, Genetically Modified , Gene Targeting , Genome , RNA, Guide, Kinetoplastida/genetics
11.
Genes Cells ; 19(7): 555-64, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24848337

ABSTRACT

The type II clustered regularly interspaced short palindromic repeats (CRISPR)/CRISPR-associated (Cas) system, which is an adaptive immune system of bacteria, has become a powerful tool for genome editing in various model organisms. Here, we demonstrate multiple genome modifications mediated by CRISPR/Cas9 in zebrafish (Danio rerio). Multiple genes including golden/gol and tyrosinase/tyr, which are involved in pigment formation, and s1pr2 and spns2, which are involved in cardiac development, were disrupted with insertion and/or deletion (indel) mutations introduced by the co-injection of multiple guide RNAs (gRNAs) and the nuclease Cas9 mRNA. We simultaneously observed two distinct phenotypes, such as, the two hearts phenotype and the hypopigmentation of skin melanophores and the retinal pigment epithelium, in the injected F0 embryos. Additionally, we detected the targeted deletion and inversion genes as a 7.1-kb fragment between the two distinct spns2 targeted sites together with indel mutations. Conversely, chromosomal translocations among five target loci were not detected. Therefore, we confirmed that the CRISPR/Cas9-induced indel mutations and a locus-specific deletion were heritable in F1 embryos. To screen founders, we improved heteroduplex mobility assay (HMA) for simultaneously detecting indel mutations in different target loci. The results suggest that the multi-locus HMA is a powerful tool for identification of multiple genome modifications mediated by the CRISPR/Cas9 system.


Subject(s)
CRISPR-Cas Systems , Genome , INDEL Mutation , Zebrafish Proteins/genetics , Zebrafish/genetics , Animals , Embryo, Nonmammalian , Genetic Engineering/methods , Genetic Loci , Heart/embryology , Inverted Repeat Sequences , Phenotype , Pigmentation/genetics , Retinal Pigment Epithelium/embryology , Zebrafish/embryology
12.
Dev Growth Differ ; 56(1): 26-33, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24117409

ABSTRACT

Zebrafish is a model vertebrate suitable for genetic analysis. Forward genetic analysis via chemical mutagenesis screening has established a variety of zebrafish mutants that are defective in various types of organogenesis, and the genes responsible for the individual mutants have been identified from genome mapping. On the other hand, reverse genetic analysis via targeted gene disruption using embryonic stem (ES) cells (e.g., knockout mouse) can uncover gene functions by investigating the phenotypic effects. However, this approach is mostly limited to mice among the vertebrate models because of the difficulty in establishing ES cells. Recently, new gene targeting technologies, such as the transcription activator-like effector nucleases (TALEN) and clustered regularly interspaced short palindromic repeats (CRISPR)/Cas9 systems, have been developed: that can directly introduce genome modifications at the targeted genomic locus. Here, we summarize these new and powerful genome editing techniques for the study of zebrafish.


Subject(s)
DNA Restriction Enzymes/metabolism , Genetic Engineering/methods , Genome/genetics , Zebrafish/genetics , Animals , CRISPR-Cas Systems , DNA Restriction Enzymes/genetics , Substrate Specificity
13.
Biochim Biophys Acta ; 1841(5): 759-65, 2014 May.
Article in English | MEDLINE | ID: mdl-23921254

ABSTRACT

Sphingosine 1-phosphate (S1P) is a lipid mediator that plays important roles in diverse cellular functions such as cell proliferation, differentiation and migration. S1P is synthesized inside the cells and subsequently released to the extracellular space, where it binds to specific receptors that are located on the plasma membranes of target cells. Accumulating recent evidence suggests that S1P transporters including SPNS2 mediate S1P release from the cells and are involved in the physiological functions of S1P. In this review, we discuss recent advances in our understanding of the mechanism and physiological functions of S1P transporters. This article is part of a Special Issue entitled New Frontiers in Sphingolipid Biology.


Subject(s)
Anion Transport Proteins/metabolism , Cell Physiological Phenomena , Lysophospholipids/metabolism , Sphingosine/analogs & derivatives , Animals , Humans , Sphingosine/metabolism
14.
Biol Open ; 2(8): 789-94, 2013 Aug 15.
Article in English | MEDLINE | ID: mdl-23951404

ABSTRACT

The lipid mediator sphingosine-1-phosphate (S1P) is a regulator of cardiac development in zebrafish, as disruption of its receptor s1pr2 or transporter spns2 causes migration defects in cardiac progenitors. To examine the genetic interaction of S1P signaling and the cell adhesion molecule fibronectin, we have established a fn;spns2 double mutant. Cardiac migration defects in fn;spns2 mutants were more severe than those in fn or spns2 mutants. We further found that the lower jaw morphology was disorganized in the fn;spns2 mutant, while it had a slightly shortened anterior-posterior distance in the ventral pharyngeal arch in fn and spns2 mutants relative to wild type. Knockdown of fn in the s1pr2 mutant, but not in the s1pr1 mutant, resulted in severe defects in cardiac migration and ventral pharyngeal arch arrangement. Further, in the background of the fn mutant, knockdown of endothelin receptor A (ednra), which was downregulated in the spns2 mutant, caused pharyngeal defects resembling those in the fn;spns2 mutant. These results strongly suggest that Spns2-S1PR2 signaling and fibronectin cooperatively regulate both cardiac and lower jaw development in zebrafish.

15.
Genes Cells ; 18(6): 450-8, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23573916

ABSTRACT

The heteroduplex mobility assay (HMA) is widely used to characterize strain variants of human viruses. To determine whether it can detect small sequence differences in homologous templates, we constructed a series of deletion constructs (1-10 bp deletions) in the multiple cloning site (MCS) of pBluescript II. After PCR amplification of the MCS using a mixture of wild-type and one of the deletion constructs, the resulting PCR amplicons were electrophoresed using 15% polyacrylamide gels. Two types of heteroduplexes exhibited retarded electrophoretic migration compared with individual homoduplexes. Therefore, we applied this HMA to detect transcription activator-like effector nucleases (TALEN)-induced insertion and/or deletion (indel) mutations at an endogenous locus. We found that TALEN in vivo activity was easily estimated by the degree of multiple HMA profiles derived from TALEN-injected F0 embryos. Furthermore, TALEN-injected F0 founder fish produced several unique HMA profiles in F1 embryos. Sequence analysis confirmed that the different HMA profiles contained distinct indel mutations. Thus, HMA is a rapid and sensitive analytical method for the detection of the TALEN-mediated genome modifications.


Subject(s)
Deoxyribonucleases/metabolism , Genome/genetics , Heteroduplex Analysis , Zebrafish/genetics , Animals , INDEL Mutation/genetics
16.
Biol Open ; 2(4): 363-7, 2013 Apr 15.
Article in English | MEDLINE | ID: mdl-23616919

ABSTRACT

Artificially designed nucleases such as zinc-finger nucleases (ZFNs) and transcription activator-like effector nucleases (TALENs) can induce a targeted DNA double-strand break at the specific target genomic locus, leading to the frameshift-mediated gene disruption. However, the assays for their activity on the endogenous genomic loci remain limited. Herein, we describe a versatile modified lacZ assay to detect frameshifts in the nuclease target site. Short fragments of the genome DNA at the target or putative off-target loci were amplified from the genomic DNA of TALEN-treated or control embryos, and were inserted into the lacZα sequence for the conventional blue-white selection. The frequency of the frameshifts in the fragment can be estimated from the numbers of blue and white colonies. Insertions and/or deletions were easily determined by sequencing the plasmid DNAs recovered from the positive colonies. Our technique should offer broad application to the artificial nucleases for genome editing in various types of model organisms.

17.
J Biochem ; 152(4): 305-11, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22923732

ABSTRACT

The lipid mediator sphingosine-1-phosphate (S1P) is generated within cells from sphingosine by two sphingosine kinases (SPHK1 and SPHK2). Intracellularly synthesized S1P is released into the extracellular fluid by S1P transporters, including SPNS2. Released S1P binds specifically to the G protein-coupled S1P receptors (S1PR1/S1P(1)-S1PR5/S1P(5)), which activate a diverse range of downstream signalling pathways. Recent studies have proposed that one of the central physiological functions of intercellular S1P signalling is in lymphocyte trafficking in vivo because genetic disruption of SPHK1/2, SPNS2 or S1PR1/S1P(1) in mice induces a lymphopenia phenotype. In this review, we discuss the current understanding of intercellular S1P signalling in the context of immunity.


Subject(s)
Immunity , Lysophospholipids/immunology , Sphingosine/analogs & derivatives , Animals , Humans , Lysophospholipids/metabolism , Signal Transduction , Sphingosine/immunology , Sphingosine/metabolism
18.
PLoS One ; 7(6): e38941, 2012.
Article in English | MEDLINE | ID: mdl-22723910

ABSTRACT

Sphingosine-1-phosphate (S1P), a sphingolipid metabolite that is produced inside the cells, regulates a variety of physiological and pathological responses via S1P receptors (S1P1-5). Signal transduction between cells consists of three steps; the synthesis of signaling molecules, their export to the extracellular space and their recognition by receptors. An S1P concentration gradient is essential for the migration of various cell types that express S1P receptors, such as lymphocytes, pre-osteoclasts, cancer cells and endothelial cells. To maintain this concentration gradient, plasma S1P concentration must be at a higher level. However, little is known about the molecular mechanism by which S1P is supplied to extracellular environments such as blood plasma. Here, we show that SPNS2 functions as an S1P transporter in vascular endothelial cells but not in erythrocytes and platelets. Moreover, the plasma S1P concentration of SPNS2-deficient mice was reduced to approximately 60% of wild-type, and SPNS2-deficient mice were lymphopenic. Our results demonstrate that SPNS2 is the first physiological S1P transporter in mammals and is a key determinant of lymphocyte egress from the thymus.


Subject(s)
Anion Transport Proteins/metabolism , Endothelial Cells/metabolism , Lysophospholipids/metabolism , Sphingosine/analogs & derivatives , Animals , Anion Transport Proteins/deficiency , Anion Transport Proteins/genetics , Blood Platelets/metabolism , Erythrocytes/metabolism , Female , Gene Targeting , Humans , Lymphocytes/metabolism , Lysophospholipids/blood , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Sphingosine/blood , Sphingosine/metabolism , Thymus Gland/metabolism , Transcription, Genetic
19.
J Biol Chem ; 286(3): 1758-66, 2011 Jan 21.
Article in English | MEDLINE | ID: mdl-21084291

ABSTRACT

FTY720 is a novel immunomodulating drug that can be phosphorylated inside cells; its phosphorylated form, FTY720-P, binds to a sphingosine 1-phosphate (S1P) receptor, S1P(1), and inhibits lymphocyte egress into the circulating blood. Although the importance of its pharmacological action has been well recognized, little is known about how FTY720-P is released from cells after its phosphorylation inside cells. Previously, we showed that zebrafish Spns2 can act as an S1P exporter from cells and is essential for zebrafish heart formation. Here, we demonstrate that human SPNS2 can transport several S1P analogues, including FTY720-P. Moreover, FTY720-P is transported by SPNS2 through the same pathway as S1P. This is the first identification of an FTY720-P transporter in cells; this finding is important for understanding FTY720 metabolism.


Subject(s)
Carrier Proteins/metabolism , Immunosuppressive Agents/pharmacology , Membrane Proteins/metabolism , Propylene Glycols/pharmacology , Sphingosine/analogs & derivatives , Animals , Biological Transport/drug effects , Biological Transport/physiology , CHO Cells , Carrier Proteins/genetics , Cricetinae , Cricetulus , Fingolimod Hydrochloride , Humans , Immunosuppressive Agents/pharmacokinetics , Lysophospholipids/genetics , Lysophospholipids/metabolism , Membrane Proteins/genetics , Mice , Phosphorylation/drug effects , Phosphorylation/physiology , Propylene Glycols/pharmacokinetics , Receptors, Lysosphingolipid/genetics , Receptors, Lysosphingolipid/metabolism , Sphingosine/genetics , Sphingosine/metabolism , Sphingosine/pharmacokinetics , Sphingosine/pharmacology
20.
Science ; 323(5913): 524-7, 2009 Jan 23.
Article in English | MEDLINE | ID: mdl-19074308

ABSTRACT

Sphingosine-1-phosphate (S1P) is a secreted lipid mediator that functions in vascular development; however, it remains unclear how S1P secretion is regulated during embryogenesis. We identified a zebrafish mutant, ko157, that displays cardia bifida (two hearts) resembling that in the S1P receptor-2 mutant. A migration defect of myocardial precursors in the ko157 mutant is due to a mutation in a multipass transmembrane protein, Spns2, and can be rescued by S1P injection. We show that the export of S1P from cells requires Spns2. spns2 is expressed in the extraembryonic tissue yolk syncytial layer (YSL), and the introduction of spns2 mRNA in the YSL restored the cardiac defect in the ko157 mutant. Thus, Spns2 in the YSL functions as a S1P transporter in S1P secretion, thereby regulating myocardial precursor migration.


Subject(s)
Carrier Proteins/metabolism , Embryo, Nonmammalian/metabolism , Heart/embryology , Lysophospholipids/metabolism , Membrane Proteins/metabolism , Sphingosine/analogs & derivatives , Zebrafish Proteins/metabolism , Zebrafish/embryology , Animals , Animals, Genetically Modified , Blastomeres/metabolism , CHO Cells , Carrier Proteins/chemistry , Carrier Proteins/genetics , Cell Movement , Cricetinae , Cricetulus , Embryo, Nonmammalian/cytology , Embryonic Development , Heart Defects, Congenital/embryology , Humans , Membrane Proteins/chemistry , Membrane Proteins/genetics , Mesoderm/metabolism , Mice , Molecular Sequence Data , Mutation , Oligonucleotides, Antisense , Organogenesis , Signal Transduction , Somites/embryology , Somites/metabolism , Sphingosine/metabolism , Zebrafish/genetics , Zebrafish Proteins/chemistry , Zebrafish Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...