Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
1.
J Lipid Res ; 57(11): 2088-2094, 2016 11.
Artigo em Inglês | MEDLINE | ID: mdl-27655910

RESUMO

Sphingosine-1-phosphate (S1P) is present in the blood plasma and acts as a pivotal intercellular signal transmitter in the immune system by recruiting lymphocytes from the thymus and secondary lymphoid tissues. The plasma S1P concentration is maintained by the supply of S1P from erythrocytes. Previously, we showed that S1P release from erythrocytes is mediated by an ATP-dependent transporter. In this study, we attempted to establish a rapid and reliable method for measuring the S1P transport activity in erythrocytes by using a fluorescent S1P analog, 7-nitro-2-1,3-benzoxadiazol-4-yl (NBD)-labeled S1P. NBD-S1P was released from erythrocytes in a time-dependent manner. The NBD-S1P release was reduced after exposure to glyburide, which is an inhibitor of the S1P transporter in erythrocytes. Moreover, the release of NBD-S1P and S1P from erythrocytes was competitively inhibited by intracellular S1P and NBD-S1P, respectively. These results showed that the erythrocyte S1P transporter exports NBD-S1P. We optimized the sample-preparation conditions and lipid extraction to increase the sensitivity of the assay. Furthermore, we successfully measured NBD-S1P release without lipid extraction by decreasing the concentration of BSA in the assay buffer to 0.1%. This method will be useful for the high-throughput screening of S1P transporter inhibitors using conventional fluorometers.


Assuntos
Plaquetas/química , Eritrócitos/química , Lisofosfolipídeos/isolamento & purificação , Esfingosina/análogos & derivados , Fluorescência , Humanos , Lisofosfolipídeos/sangue , Lisofosfolipídeos/química , Esfingosina/sangue , Esfingosina/química , Esfingosina/isolamento & purificação
2.
Biochim Biophys Acta ; 1841(5): 759-65, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-23921254

RESUMO

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.


Assuntos
Proteínas de Transporte de Ânions/metabolismo , Fenômenos Fisiológicos Celulares , Lisofosfolipídeos/metabolismo , Esfingosina/análogos & derivados , Animais , Humanos , Esfingosina/metabolismo
3.
J Biol Chem ; 286(3): 1758-66, 2011 Jan 21.
Artigo em Inglês | MEDLINE | ID: mdl-21084291

RESUMO

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.


Assuntos
Proteínas de Transporte/metabolismo , Imunossupressores/farmacologia , Proteínas de Membrana/metabolismo , Propilenoglicóis/farmacologia , Esfingosina/análogos & derivados , Animais , Transporte Biológico/efeitos dos fármacos , Transporte Biológico/fisiologia , Células CHO , Proteínas de Transporte/genética , Cricetinae , Cricetulus , Cloridrato de Fingolimode , Humanos , Imunossupressores/farmacocinética , Lisofosfolipídeos/genética , Lisofosfolipídeos/metabolismo , Proteínas de Membrana/genética , Camundongos , Fosforilação/efeitos dos fármacos , Fosforilação/fisiologia , Propilenoglicóis/farmacocinética , Receptores de Lisoesfingolipídeo/genética , Receptores de Lisoesfingolipídeo/metabolismo , Esfingosina/genética , Esfingosina/metabolismo , Esfingosina/farmacocinética , Esfingosina/farmacologia
4.
Angew Chem Int Ed Engl ; 51(2): 509-12, 2012 Jan 09.
Artigo em Inglês | MEDLINE | ID: mdl-22105970

RESUMO

Fluorescent combination: Cell-penetrating probes derived from the diterpene fusicoccin can form ternary complexes with 14-3-3 proteins and phosphopeptide ligands, whereupon the probes site-specifically attach a fluorescent tag onto the surface of the 14-3-3 proteins.


Assuntos
Proteínas 14-3-3/análise , Corantes Fluorescentes/química , Glicosídeos/química , Fosfopeptídeos/química , Proteínas 14-3-3/química , Linhagem Celular , Humanos , Modelos Moleculares , Coloração e Rotulagem
5.
Sci Rep ; 12(1): 21603, 2022 Dec 14.
Artigo em Inglês | MEDLINE | ID: mdl-36517530

RESUMO

This report presents n-type single-walled carbon nanotubes (SWCNT) films with ultra-long air stability using a cationic surfactant and demonstrates that the n-type Seebeck coefficient can be maintained for more than two years, which is the highest stability reported thus far to the best of our knowledge. Furthermore, the SWCNT films exhibit an extremely low thermal conductivity of 0.62 ± 0.08 W/(m·K) in the in-plane direction, which is very useful for thin-film TEGs. We fabricated all-carbon-nanotube TEGs, which use p-type SWCNT films and the n-type SWCNT films developed, and their air-stability was investigated. The TEGs did not degrade for 160 days and exhibited an output voltage of 24 mV, with a maximum power of 0.4 µW at a temperature difference of 60 K. These results open a pathway to enable the widespread use of carbon nanotube TEGs as power sources in IoT sensors.

6.
Biochem Biophys Res Commun ; 395(3): 387-94, 2010 May 07.
Artigo em Inglês | MEDLINE | ID: mdl-20382126

RESUMO

OBJECTIVES: To determine the role of macrophage ATP-binding cassette transporter A5 (ABCA5) in cellular cholesterol homeostasis and atherosclerotic lesion development. METHODS AND RESULTS: Chimeras with dysfunctional macrophage ABCA5 (ABCA5(-M/-M)) were generated by transplantation of bone marrow from ABCA5 knockout (ABCA5(-/-)) mice into irradiated LDLr(-/-) mice. In vitro, bone marrow-derived macrophages from ABCA5(-M/-M) chimeras exhibited a 29% (P<0.001) decrease in cholesterol efflux to HDL, whereas a 21% (P=0.07) increase in cholesterol efflux to apoA-I was observed. Interestingly, expression of ABCA1, but not ABCG1, was up-regulated in absence of functional ABCA5 in macrophages. To induce atherosclerosis, the transplanted LDLr(-/-) mice were fed a high-cholesterol Western-type diet (WTD) for 6, 10, or 18weeks, allowing analysis of effects on initial as well as advanced lesion development. Atherosclerosis development was not affected in male ABCA5(-M/-M) chimeras after 6, 10, and 18weeks WTD feeding. However, female ABCA5(-M/-M) chimeras did develop significantly (P<0.05) larger aortic root lesions as compared with female controls after 6 and 10weeks WTD feeding. CONCLUSIONS: ABCA5 influences macrophage cholesterol efflux, and selective disruption of ABCA5 in macrophages leads to increased atherosclerotic lesion development in female LDLr(-/-) mice.


Assuntos
Transportadores de Cassetes de Ligação de ATP/metabolismo , Aterosclerose/metabolismo , Colesterol/metabolismo , Macrófagos/metabolismo , Transportadores de Cassetes de Ligação de ATP/genética , Animais , Colesterol/genética , Feminino , Lipídeos/sangue , Masculino , Camundongos , Camundongos Knockout , Receptores de LDL/genética
7.
Materials (Basel) ; 13(21)2020 Oct 29.
Artigo em Inglês | MEDLINE | ID: mdl-33138219

RESUMO

A spot periodic heating method is a highly accurate, non-contact method for the evaluation of anisotropy and relative thermophysical property distribution. However, accurately evaluating thermal diffusivity is difficult due to the influence of temperature wave reflection from the whole surface of the sample. This study proposes a method to derive thermal diffusivity using a parameter table based on heat transfer equations using the concept of optimum distance between heating-point and measurement point. This method considers finite sample size, sensitivity distribution of infrared ray detector, intensity distribution of heating laser and sample thickness. In these results, the obtained thermal diffusivity of pure copper corresponded well with previous literature values. In conclusion, this method is considered highly effective in evaluating the thermal diffusivity in the horizontal direction.

8.
Cells ; 8(1)2019 01 10.
Artigo em Inglês | MEDLINE | ID: mdl-30634695

RESUMO

Broad-spectrum drug resistance is a major obstacle in cancer treatment, which is often caused by overexpression of ABC transporters the levels of which vary between individuals due to single-nucleotide polymorphisms (SNPs) in their genes. In the present study, we focused on the human ABC transporter ABCC4 and one major non-synonymous SNP variant of the ABCC4 gene in the Japanese population (rs11568658, 559 G > T, G187W) whose allele frequency is 12.5%. Cells expressing ABCC4 (G187W) were established using the Flp-In™ system based on Flp recombinase-mediated transfection to quantitatively evaluate the impacts of this non-synonymous SNP on drug resistance profiles of the cells. Cells expressing ABCC4 (WT) or (G187W) showed comparable ABCC4 mRNA levels. 3-(4,5-Dimethyl-2-thiazol-2-yl)-2,5-diphenyl-2H-tetrazolium bromide (MTT) assay indicated that the EC50 value of the anticancer drug, SN-38, against cells expressing ABCC4 (G187W) was 1.84-fold lower than that against cells expressing ABCC4 (WT). Both azathioprine and 6-mercaptopurine showed comparable EC50 values against cells expressing ABCC4 (G187W) and those expressing ABCC4 (WT). These results indicate that the substitution of Gly at position 187 of ABCC4 to Trp resulted in reduced SN-38 resistance.


Assuntos
Antineoplásicos/farmacologia , Resistencia a Medicamentos Antineoplásicos/genética , Irinotecano/farmacologia , Proteínas Associadas à Resistência a Múltiplos Medicamentos/genética , Células HEK293 , Humanos , Polimorfismo de Nucleotídeo Único
9.
Mol Cell Biol ; 25(10): 4138-49, 2005 May.
Artigo em Inglês | MEDLINE | ID: mdl-15870284

RESUMO

ABCA5 is a member of the ABC transporter A subfamily, and a mouse orthologue (mABCA5) in newborn mouse brain and neural cells was identified by reverse transcription-PCR. Full-length cDNA cloning revealed that mABCA5 consists of 1,642 amino acid residues and that its putative structure is that of a full-type ABC transporter having two sets of six transmembrane segments and a nucleotide binding domain. Immunohistochemical studies revealed that mABCA5 is expressed in brain, lung, heart, and thyroid gland. A subcellular localization analysis showed that mABCA5 is a resident of lysosomes and late endosomes. Abca5(-)(/)(-) mice exhibited symptoms similar to those of several lysosomal diseases in heart, although no prominent abnormalities were found in brain or lung. They developed a dilated cardiomyopathy-like heart after reaching adulthood and died due to depression of the cardiovascular system. In addition, Abca5(-)(/)(-) mice also exhibited exophthalmos and collapse of the thyroid gland. Therefore, ABCA5 is a protein related to a lysosomal disease and plays important roles, especially in cardiomyocytes and follicular cells.


Assuntos
Transportadores de Cassetes de Ligação de ATP/genética , Transportadores de Cassetes de Ligação de ATP/metabolismo , Lisossomos/metabolismo , Lisossomos/patologia , Transportadores de Cassetes de Ligação de ATP/química , Anormalidades Múltiplas/genética , Anormalidades Múltiplas/metabolismo , Anormalidades Múltiplas/patologia , Sequência de Aminoácidos , Animais , Linhagem Celular , Clonagem Molecular , Exoftalmia/genética , Exoftalmia/metabolismo , Exoftalmia/patologia , Feminino , Deleção de Genes , Perfilação da Expressão Gênica , Humanos , Imuno-Histoquímica , Masculino , Camundongos , Camundongos Knockout , Dados de Sequência Molecular , Miocárdio/metabolismo , Miocárdio/patologia , Transporte Proteico , RNA Mensageiro/análise , RNA Mensageiro/genética , Ratos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Glândula Tireoide/metabolismo , Hormônios Tireóideos/metabolismo
10.
Methods Mol Biol ; 1697: 73-82, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-28456950

RESUMO

Sphingosine-1-phosphate (S1P) is an intercellular signaling molecule that is present in the plasma and plays an important role in recruiting lymphocytes from the thymus and secondary lymphoid organs. Erythrocytes are the most abundant cells in the blood and substantially contribute to the S1P supply in the plasma by releasing intracellularly synthesized S1P via an S1P transporter. Thus, the S1P transporter in erythrocytes is a potential target for immuno-suppressing drugs.In this chapter, we describe a rapid method for measuring the activity of the erythrocyte S1P transporter by using the fluorescent S1P analog, 7-nitro-2-1,3-benzoxadiazol-4-yl (NBD)-labeled S1P. This method does not require chromatography performed with high-performance liquid chromatography, liquid chromatography-tandem mass spectrometry, or thin-layer chromatography methods. Furthermore, S1P transporter activity can be detected by measuring the increase in fluorescence intensity in the extracellular buffer without performing lipid extraction.


Assuntos
Eritrócitos/química , Proteínas de Transporte de Fosfato/metabolismo , Animais , Transporte Biológico , Eritrócitos/metabolismo , Fluorescência , Humanos , Lisofosfolipídeos/metabolismo , Esfingosina/análogos & derivados , Esfingosina/metabolismo
11.
Sci Rep ; 8(1): 4969, 2018 03 21.
Artigo em Inglês | MEDLINE | ID: mdl-29563527

RESUMO

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.


Assuntos
Células Eritroides/metabolismo , Lisofosfolipídeos/metabolismo , Proteínas de Membrana/metabolismo , Esfingosina/análogos & derivados , Animais , Células CHO , Linhagem Celular , Cricetulus , Técnicas de Inativação de Genes , Proteínas de Membrana/genética , Camundongos , Análise em Microsséries , Fosfotransferases (Aceptor do Grupo Álcool)/genética , Fosfotransferases (Aceptor do Grupo Álcool)/metabolismo , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Esfingosina/metabolismo
12.
Biochem Biophys Res Commun ; 364(4): 1032-6, 2007 Dec 28.
Artigo em Inglês | MEDLINE | ID: mdl-17971301

RESUMO

We have identified splicing variants of the mouse a4 subunit which have the same open reading frame but have a different 5'-noncoding sequence. Further determination of the 5'-upstream region of the a4 gene in mouse indicated the presence of two first exons (exon 1a and exon 1b) which include the 5'-noncoding sequence of each variant. The mRNAs of both splicing variants (a4-I and a4-II) show a similar expression pattern in mouse kidney by in situ hybridization. However, tissue and developmental expression patterns of the variants are different. In addition to strong expression in kidney, a4-I expression was detected in heart, lung, skeletal muscle, and testis, whereas a4-II is expressed in lung, liver, and testis. During development, a4-I was expressed beginning with the early embryonic stage, but a4-II mRNA was detected from day 17. These results suggest that each a4 variant has both a tissue and developmental stage specific function.


Assuntos
Rim/metabolismo , Especificidade de Órgãos/genética , Isoformas de Proteínas/genética , ATPases Vacuolares Próton-Translocadoras/genética , ATPases Vacuolares Próton-Translocadoras/metabolismo , Animais , Camundongos , Distribuição Tecidual
13.
Biochim Biophys Acta ; 1555(1-3): 71-4, 2002 Sep 10.
Artigo em Inglês | MEDLINE | ID: mdl-12206894

RESUMO

The vacuolar (H(+))-ATPases (or V-ATPases) are ATP-dependent proton pumps that function to acidify intracellular compartments in eukaryotic cells. This acidification is essential for such processes as receptor-mediated endocytosis, intracellular targeting of lysosomal enzymes, protein processing and degradation and the coupled transport of small molecules. V-ATPases in the plasma membrane of specialized cells also function in such processes as renal acidification, bone resorption and pH homeostasis. Work from our laboratory has focused on the V-ATPases from clathrin-coated vesicles and yeast vacuoles.Structurally, the V-ATPases are composed of two domains: a peripheral complex (V(1)) composed of eight different subunits (A-H) that is responsible for ATP hydrolysis and an integral complex (V(0)) composed of five different subunits (a, d, c, c' and c") that is responsible for proton translocation. Electron microscopy has revealed the presence of multiple stalks connecting the V(1) and V(0) domains, and crosslinking has been used to address the arrangement of subunits in the complex. Site-directed mutagenesis has been employed to identify residues involved in ATP hydrolysis and proton translocation and to study the topology of the 100 kDa a subunit. This subunit has been shown to control intracellular targeting of the V-ATPase and to influence reversible dissociation and coupling of proton transport and ATP hydrolysis.


Assuntos
Vesículas Revestidas por Clatrina/enzimologia , ATPases Translocadoras de Prótons/metabolismo , Leveduras/enzimologia , Trifosfato de Adenosina/química , Trifosfato de Adenosina/metabolismo , Modelos Moleculares , ATPases Translocadoras de Prótons/química , ATPases Translocadoras de Prótons/genética
14.
FEBS Lett ; 545(1): 76-85, 2003 Jun 12.
Artigo em Inglês | MEDLINE | ID: mdl-12788495

RESUMO

The vacuolar H(+)-ATPases (or V-ATPases) are a family of ATP-dependent proton pumps responsible for acidification of intracellular compartments and, in certain cases, proton transport across the plasma membrane of eukaryotic cells. They are multisubunit complexes composed of a peripheral domain (V(1)) responsible for ATP hydrolysis and an integral domain (V(0)) responsible for proton translocation. Based upon their structural similarity to the F(1)F(0) ATP synthases, the V-ATPases are thought to operate by a rotary mechanism in which ATP hydrolysis in V(1) drives rotation of a ring of proteolipid subunits in V(0). This review is focused on the current structural knowledge of the V-ATPases as it relates to the mechanism of ATP-driven proton translocation.


Assuntos
Trifosfato de Adenosina/metabolismo , Prótons , ATPases Vacuolares Próton-Translocadoras/química , ATPases Vacuolares Próton-Translocadoras/metabolismo , Nucleotídeos de Adenina/metabolismo , Domínio Catalítico , Hidrólise , Transporte de Íons , Modelos Biológicos , Subunidades Proteicas , ATPases Translocadoras de Prótons/química , ATPases Vacuolares Próton-Translocadoras/ultraestrutura
15.
J Biochem ; 152(4): 305-11, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22923732

RESUMO

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.


Assuntos
Imunidade , Lisofosfolipídeos/imunologia , Esfingosina/análogos & derivados , Animais , Humanos , Lisofosfolipídeos/metabolismo , Transdução de Sinais , Esfingosina/imunologia , Esfingosina/metabolismo
16.
PLoS One ; 7(6): e38941, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22723910

RESUMO

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.


Assuntos
Proteínas de Transporte de Ânions/metabolismo , Células Endoteliais/metabolismo , Lisofosfolipídeos/metabolismo , Esfingosina/análogos & derivados , Animais , Proteínas de Transporte de Ânions/deficiência , Proteínas de Transporte de Ânions/genética , Plaquetas/metabolismo , Eritrócitos/metabolismo , Feminino , Marcação de Genes , Humanos , Linfócitos/metabolismo , Lisofosfolipídeos/sangue , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Esfingosina/sangue , Esfingosina/metabolismo , Timo/metabolismo , Transcrição Gênica
19.
J Biol Chem ; 284(32): 21192-200, 2009 Aug 07.
Artigo em Inglês | MEDLINE | ID: mdl-19531471

RESUMO

Sphingosine 1-phosphate (S1P) is a bioactive lipid signal transmitter present in blood. Blood plasma S1P is supplied from erythrocytes and plays an important role in lymphocyte egress from lymphoid organs. However, the S1P export mechanism from erythrocytes to blood plasma is not well defined. To elucidate the mechanism of S1P export from erythrocytes, we performed the enzymatic characterization of S1P transporter in rat erythrocytes. Rat erythrocytes constitutively released S1P without any stimulus. The S1P release was reduced by an ABCA1 transporter inhibitor, glyburide, but not by a multidrug resistance-associated protein inhibitor, MK571, or a multidrug resistance protein inhibitor, cyclosporine A. Furthermore, we measured S1P transport activity using rat erythrocyte inside-out membrane vesicles (IOVs). Although the effective S1P transport into IOVs was observed in the presence of ATP, this activity was also supported by dATP and adenosine 5'-(beta,gamma-imido)triphosphate. The rate of S1P transport increased depending on S1P concentration, with an apparent K(m) value of 21 microm. Two phosphorylated sphingolipids, dihydrosphingosine 1-phosphate and ceramide 1-phosphate, did not inhibit S1P transport. Similar to the intact erythrocytes, the uptake of S1P into IOVs was inhibited by glyburide and vanadate but not by the other ABC transporter inhibitors. These results suggest that S1P is exported from the erythrocytes by a novel ATP-dependent transporter.


Assuntos
Trifosfato de Adenosina/metabolismo , Eritrócitos/metabolismo , Lisofosfolipídeos/química , Proteínas Associadas à Resistência a Múltiplos Medicamentos/química , Esfingosina/análogos & derivados , Animais , Transporte Biológico , Ciclosporina/metabolismo , Relação Dose-Resposta a Droga , Feminino , Glibureto/química , Cinética , Lisofosfolipídeos/metabolismo , Modelos Biológicos , Proteínas Associadas à Resistência a Múltiplos Medicamentos/fisiologia , Propionatos/farmacologia , Quinolinas/farmacologia , Ratos , Ratos Wistar , Esfingosina/química , Esfingosina/metabolismo , Vanadatos/química
20.
Science ; 323(5913): 524-7, 2009 Jan 23.
Artigo em Inglês | MEDLINE | ID: mdl-19074308

RESUMO

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.


Assuntos
Proteínas de Transporte/metabolismo , Embrião não Mamífero/metabolismo , Coração/embriologia , Lisofosfolipídeos/metabolismo , Proteínas de Membrana/metabolismo , Esfingosina/análogos & derivados , Proteínas de Peixe-Zebra/metabolismo , Peixe-Zebra/embriologia , Animais , Animais Geneticamente Modificados , Blastômeros/metabolismo , Células CHO , Proteínas de Transporte/química , Proteínas de Transporte/genética , Movimento Celular , Cricetinae , Cricetulus , Embrião não Mamífero/citologia , Desenvolvimento Embrionário , Cardiopatias Congênitas/embriologia , Humanos , Proteínas de Membrana/química , Proteínas de Membrana/genética , Mesoderma/metabolismo , Camundongos , Dados de Sequência Molecular , Mutação , Oligonucleotídeos Antissenso , Organogênese , Transdução de Sinais , Somitos/embriologia , Somitos/metabolismo , Esfingosina/metabolismo , Peixe-Zebra/genética , Proteínas de Peixe-Zebra/química , Proteínas de Peixe-Zebra/genética
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa