Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
1.
J Biol Chem ; 291(53): 27343-27353, 2016 12 30.
Article in English | MEDLINE | ID: mdl-27856637

ABSTRACT

Previously, we showed that levels of sphingosine-1 phosphate receptor 3 (S1PR3) are increased in a panel of cultured human lung adenocarcinoma cell lines, and that S1PR3-mediated signaling pathways regulate proliferation, soft agar growth, and invasion of human lung adenocarcinoma cells in vitro In the present study, we examine S1PR3 levels in human lung adenocarcinoma specimens. cDNA array and tumor microarray analysis shows that mRNA and protein levels of S1PR3 are significantly increased in human lung adenocarcinomas when compared with normal lung epithelial cells. Promoter analysis shows 16 candidate SMAD3 binding sites in the promoter region of S1PR3. ChIP indicates that TGF-ß treatment stimulates the binding of SMAD3 to the promoter region of S1PR3. Luciferase reporter assay demonstrates that SMAD3 transactivates S1PR3 promoter. TGF-ß stimulation or ectopic expression of TGF-ß up-regulates S1PR3 levels in vitro and ex vivo Pharmacologic inhibition of TGF-ß receptor or SMAD3 abrogates the TGF-ß-stimulated S1PR3 up-regulation. Moreover, S1PR3 knockdown dramatically inhibits tumor growth and lung metastasis, whereas ectopic expression of S1PR3 promotes the growth of human lung adenocarcinoma cells in animals. Pharmacological inhibition of S1PR3 profoundly inhibits the growth of lung carcinoma in mice. Our studies suggest that levels of S1PR3 are up-regulated in human lung adenocarcinomas, at least in part due to the TGF-ß/SMAD3 signaling axis. Furthermore, S1PR3 activity promotes the progression of human lung adenocarcinomas. Therefore, S1PR3 may represent a novel therapeutic target for the treatment of deadly lung adenocarcinomas.


Subject(s)
Adenocarcinoma/secondary , Lung Neoplasms/pathology , Receptors, Lysosphingolipid/metabolism , Smad3 Protein/metabolism , Transforming Growth Factor beta/metabolism , Adenocarcinoma/genetics , Adenocarcinoma/metabolism , Animals , Apoptosis , Blotting, Western , Cell Proliferation , Cells, Cultured , Female , Humans , Lung/metabolism , Lung/pathology , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, SCID , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Receptors, Lysosphingolipid/genetics , Reverse Transcriptase Polymerase Chain Reaction , Smad3 Protein/genetics , Sphingosine-1-Phosphate Receptors , Transforming Growth Factor beta/genetics , Xenograft Model Antitumor Assays
2.
J Biol Chem ; 291(31): 16001-10, 2016 07 29.
Article in English | MEDLINE | ID: mdl-27246851

ABSTRACT

Excessive adipocyte lipolysis generates lipid mediators and triggers inflammation in adipose tissue. However, the specific roles of lipolysis-generated mediators in adipose inflammation remain to be elucidated. In the present study, cultured 3T3-L1 adipocytes were treated with isoproterenol to activate lipolysis and the fatty acyl lipidome of released lipids was determined by using LC-MS/MS. We observed that ß-adrenergic activation elevated levels of approximately fifty lipid species, including metabolites of cyclooxygenases, lipoxygenases, epoxygenases, and other sources. Moreover, we found that ß-adrenergic activation induced cyclooxygenase 2 (COX-2), not COX-1, expression in a manner that depended on activation of hormone-sensitive lipase (HSL) in cultured adipocytes and in the epididymal white adipose tissue (EWAT) of C57BL/6 mice. We found that lipolysis activates the JNK/NFκB signaling pathway and inhibition of the JNK/NFκB axis abrogated the lipolysis-stimulated COX-2 expression. In addition, pharmacological inhibition of COX-2 activity diminished levels of COX-2 metabolites during lipolytic activation. Inhibition of COX-2 abrogated the induction of CCL2/MCP-1 expression by ß-adrenergic activation and prevented recruitment of macrophage/monocyte to adipose tissue. Collectively, our data indicate that excessive adipocyte lipolysis activates the JNK/NFκB pathway leading to the up-regulation of COX-2 expression and recruitment of inflammatory macrophages.


Subject(s)
Adipocytes/enzymology , Cyclooxygenase 2/biosynthesis , Eicosanoids/biosynthesis , Lipolysis , Panniculitis/enzymology , Signal Transduction , 3T3-L1 Cells , Adipocytes/pathology , Animals , Chemokine CCL2/metabolism , Inflammation/enzymology , Inflammation/pathology , MAP Kinase Kinase 4/metabolism , Macrophages/metabolism , Macrophages/pathology , Mice , NF-kappa B/metabolism , Panniculitis/pathology , Sterol Esterase/metabolism
3.
FASEB J ; 30(6): 2360-9, 2016 06.
Article in English | MEDLINE | ID: mdl-26965684

ABSTRACT

Previously we identified and deorphaned G-protein-coupled receptor 31 (GPR31) as the high-affinity 12(S)-hydroxyeicosatetraenoic acid [12(S)-HETE] receptor (12-HETER1). Here we have determined its distribution in prostate cancer tissue and its role in prostate tumorigenesis using in vitro and in vivo assays. Data-mining studies strongly suggest that 12-HETER1 expression positively correlates with the aggressiveness and progression of prostate tumors. This was corroborated with real-time PCR analysis of human prostate tumor tissue arrays that revealed the expression of 12-HETER1 positively correlates with the clinical stages of prostate cancers and Gleason scores. Immunohistochemistry analysis also proved that the expression of 12-HETER1 is positively correlated with the grades of prostate cancer. Knockdown of 12-HETER1 in prostate cancer cells markedly reduced colony formation and inhibited tumor growth in animals. To discover the regulatory factors, 5 candidate 12-HETER1 promoter cis elements were assayed as luciferase reporter fusions in Chinese hamster ovary (CHO) cells, where the putative cis element required for gene regulation was mapped 2 kb upstream of the 12-HETER1 transcriptional start site. The data implicate 12-HETER1 in a critical new role in the regulation of prostate cancer progression and offer a novel alternative target for therapeutic intervention.-Honn, K. V., Guo, Y., Cai, Y., Lee, M.-J., Dyson, G., Zhang, W., Tucker, S. C. 12-HETER1/GPR31, a high-affinity 12(S)-hydroxyeicosatetraenoic acid receptor, is significantly up-regulated in prostate cancer and plays a critical role in prostate cancer progression.


Subject(s)
Gene Expression Regulation, Neoplastic/physiology , Prostatic Neoplasms/metabolism , Receptors, Eicosanoid/metabolism , Receptors, G-Protein-Coupled/metabolism , Up-Regulation/physiology , Animals , Cell Line , Cloning, Molecular , Computational Biology , Cricetinae , Databases, Factual , Humans , Male , Promoter Regions, Genetic , Prostatic Neoplasms/genetics , Receptors, Eicosanoid/genetics , Receptors, G-Protein-Coupled/classification , Receptors, G-Protein-Coupled/genetics , Tissue Array Analysis , Transcriptome
4.
J Biol Chem ; 289(46): 32178-32185, 2014 Nov 14.
Article in English | MEDLINE | ID: mdl-25253697

ABSTRACT

Adipocyte lipolysis can increase the production of inflammatory cytokines such as interleukin-6 (IL-6) that promote insulin resistance. However, the mechanisms that link lipolysis with inflammation remain elusive. Acute activation of ß3-adrenergic receptors (ADRB3) triggers lipolysis and up-regulates production of IL-6 in adipocytes, and both of these effects are blocked by pharmacological inhibition of hormone-sensitive lipase. We report that stimulation of ADRB3 induces expression of sphingosine kinase 1 (SphK1) and increases sphingosine 1-phosphate production in adipocytes in a manner that also depends on hormone-sensitive lipase activity. Mechanistically, we found that adipose lipolysis-induced SphK1 up-regulation is mediated by the c-Jun N-terminal kinase (JNK)/activating protein-1 signaling pathway. Inhibition of SphK1 by sphingosine kinase inhibitor 2 diminished the ADRB3-induced IL-6 production both in vitro and in vivo. Induction of IL-6 by ADRB3 activation was suppressed by siRNA knockdown of Sphk1 in cultured adipocytes and was severely attenuated in Sphk1 null mice. Conversely, ectopic expression of SphK1 increased IL-6 expression in adipocytes. Collectively, these data demonstrate that SphK1 is a critical mediator in lipolysis-triggered inflammation in adipocytes.


Subject(s)
Adipocytes/cytology , Inflammation/metabolism , Interleukin-6/metabolism , Lipolysis , Phosphotransferases (Alcohol Group Acceptor)/metabolism , 3T3-L1 Cells , Adipocytes/metabolism , Adipose Tissue/metabolism , Adipose Tissue/pathology , Animals , MAP Kinase Kinase 4/metabolism , Male , Mice , Mice, Inbred C57BL , RNA, Small Interfering/metabolism , Signal Transduction , Sphingolipids/chemistry , Tandem Mass Spectrometry
5.
Curr Atheroscler Rep ; 17(5): 504, 2015 May.
Article in English | MEDLINE | ID: mdl-25762236

ABSTRACT

Endothelial inflammation is an important risk factor in the initiation and development of vascular disease. Therefore, signaling cascades and patho-physiological outcomes of endothelial inflammation are important questions in vascular biology. Recent studies suggest that sphingosine-1-phosphate receptor subtype 2 (S1PR2) signaling in endothelial cells (ECs) play a critical role in endothelial inflammation. For example, ECs present in atherosclerotic plaques exhibit senescence phenotype. Levels of S1PR2 are markedly increased in cultured senescent ECs and in lesion regions of atherosclerotic endothelium. Also, inflammatory cytokines and mechanical flow stress profoundly increase S1PR2 levels in ECs. Inhibition of endothelial S1PR2 signaling diminishes endothelial senescence-associated functional impairments and atherogenic stimuli-induced endothelial activation. In contrast, activation of endothelial S1PR2 stimulates the production of pro-inflammatory chemokines/cytokines and lipid mediators in ECs. In this article, we will review signaling and functions of sphingosine-1-phosphate (S1P) receptors in endothelial biology, with particular focus on endothelial S1PR2 signaling-mediated endothelial inflammation.


Subject(s)
Atherosclerosis/genetics , Endothelium, Vascular/metabolism , Inflammation/genetics , Receptors, Lysosphingolipid/genetics , Animals , Atherosclerosis/metabolism , Atherosclerosis/pathology , Cells, Cultured , Endothelium, Vascular/pathology , Humans , Inflammation/metabolism , Inflammation/pathology , Receptors, Lysosphingolipid/metabolism , Signal Transduction , Sphingosine-1-Phosphate Receptors
6.
Am J Physiol Cell Physiol ; 307(2): C169-79, 2014 Jul 15.
Article in English | MEDLINE | ID: mdl-24829496

ABSTRACT

Inflammation-induced vascular endothelial dysfunction can allow plasma proteins to cross the vascular wall, causing edema. Proteins may traverse the vascular wall through two main pathways, the paracellular and transcellular transport pathways. Paracellular transport involves changes in endothelial cell junction proteins, while transcellular transport involves caveolar transcytosis. Since both processes are associated with filamentous actin formation, the two pathways are interconnected. Therefore, it is difficult to differentiate the prevailing role of one or the other pathway during various pathologies causing an increase in vascular permeability. Using a newly developed dual-tracer probing method, we differentiated transcellular from paracellular transport during hyperfibrinogenemia (HFg), an increase in fibrinogen (Fg) content. Roles of cholesterol and sphingolipids in formation of functional caveolae were assessed using a cholesterol chelator, methyl-ß-cyclodextrin, and the de novo sphingolipid synthesis inhibitor myriocin. Fg-induced formation of functional caveolae was defined by association and colocalization of Na+-K+-ATPase and plasmalemmal vesicle-associated protein-1 with use of Förster resonance energy transfer and total internal reflection fluorescence microscopy, respectively. HFg increased permeability of the endothelial cell layer mainly through the transcellular pathway. While MßCD blocked Fg-increased transcellular and paracellular transport, myriocin affected only transcellular transport. Less pial venular leakage of albumin was observed in myriocin-treated HFg mice. HFg induced greater formation of functional caveolae, as indicated by colocalization of Na+-K+-ATPase with plasmalemmal vesicle-associated protein-1 by Förster resonance energy transfer and total internal reflection fluorescence microscopy. Our results suggest that elevated blood levels of Fg alter cerebrovascular permeability mainly by affecting caveolae-mediated transcytosis through modulation of de novo sphingolipid synthesis.


Subject(s)
Brain/blood supply , Capillary Permeability/physiology , Caveolae/metabolism , Fibrinogen/metabolism , Sphingolipids/pharmacology , Animals , Cholesterol/metabolism , Chromatography, Liquid , Endothelial Cells/drug effects , Endothelial Cells/physiology , Fibrinogen/genetics , Gene Expression Regulation , Humans , Mice , Mice, Inbred C57BL , Mice, Transgenic , Sphingolipids/metabolism , Tandem Mass Spectrometry , Transcytosis , Veins/drug effects , Veins/physiology
7.
J Biol Chem ; 288(45): 32126-32137, 2013 Nov 08.
Article in English | MEDLINE | ID: mdl-24064218

ABSTRACT

Sphingosine-1-phosphate (S1P)-regulated chemotaxis plays critical roles in various physiological and pathophysiological conditions. S1P-regulated chemotaxis is mediated by the S1P family of G-protein-coupled receptors. However, molecular details of the S1P-regulated chemotaxis are incompletely understood. Cultured human lung adenocarcinoma cell lines abundantly express S1P receptor subtype 3 (S1P3), thus providing a tractable in vitro system to characterize molecular mechanism(s) underlying the S1P3 receptor-regulated chemotactic response. S1P treatment enhances CD44 expression and induces membrane localization of CD44 polypeptides via the S1P3/Rho kinase (ROCK) signaling pathway. Knockdown of CD44 completely diminishes the S1P-stimulated chemotaxis. Promoter analysis suggests that the CD44 promoter contains binding sites of the ETS-1 (v-ets erythroblastosis virus E26 oncogene homolog 1) transcriptional factor. ChIP assay confirms that S1P treatment stimulates the binding of ETS-1 to the CD44 promoter region. Moreover, S1P induces the expression and nuclear translocation of ETS-1. Knockdown of S1P3 or inhibition of ROCK abrogates the S1P-induced ETS-1 expression. Furthermore, knockdown of ETS-1 inhibits the S1P-induced CD44 expression and cell migration. In addition, we showed that S1P3/ROCK signaling up-regulates ETS-1 via the activity of JNK. Collectively, we characterized a novel signaling axis, i.e., ROCK-JNK-ETS-1-CD44 pathway, which plays an essential role in the S1P3-regulated chemotactic response.


Subject(s)
Chemotaxis/physiology , Hyaluronan Receptors/biosynthesis , Proto-Oncogene Protein c-ets-1/metabolism , Receptors, Lysosphingolipid/metabolism , Signal Transduction/physiology , Transcription, Genetic/physiology , Up-Regulation/physiology , Cell Line, Tumor , Gene Knockdown Techniques , Humans , Hyaluronan Receptors/genetics , Lysophospholipids/genetics , Lysophospholipids/metabolism , MAP Kinase Kinase 4/genetics , MAP Kinase Kinase 4/metabolism , Proto-Oncogene Protein c-ets-1/genetics , Receptors, Lysosphingolipid/agonists , Receptors, Lysosphingolipid/genetics , Sphingosine/analogs & derivatives , Sphingosine/genetics , Sphingosine/metabolism , rho-Associated Kinases/genetics , rho-Associated Kinases/metabolism
8.
Prostaglandins Other Lipid Mediat ; 106: 62-71, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23770055

ABSTRACT

Sphingosine-1-phosphate (S1P) regulates a wide array of biological functions in endothelial cells. We previously showed that S1P receptor subtype 2 (S1P2) is significantly up-regulated in the atherosclerotic endothelium (J. Biol. Chem. 283:30363, 2008). In this study, we investigated the roles of S1P2-mediated signaling in the proinflammatory responses of endothelial cells. Treatment with tumor necrosis factor-α (TNFα), a proinflammatory cytokine, increased the expression of S1P2 receptors in endothelial cells. TNFα treatment also enhanced sphingosine kinase 1 expression and increased S1P production. Pharmacological inhibition or knockdown of S1P2 receptors completely abrogated the TNFα-induced VCAM-1 (vascular cell adhesion molecule 1) and ICAM-1 (intercellular adhesion molecule 1) expression in endothelial cells. In contrast, pharmacological inhibition or knockdown of other S1P receptor subtypes had no effect on the TNFα-stimulated ICAM-1 and VCAM-1 expression. Moreover, ectopic expression of S1P2 receptors increased VCAM-1 and ICAM-1 expression in endothelial cells in response to S1P stimulation. Mechanistically, we show that antagonizing S1P2 signaling markedly inhibited the TNFα-stimulated NFκB activation. Utilizing the NFκB reporter luciferase assay, the S1P/S1P2 signaling was shown to stimulate NFκB activation. Moreover, the S1P/S1P2-stimulated VCAM-1/ICAM-1 expression was completely abolished by the pharmacological inhibitor of NFκB. Collectively, our data suggest that TNFα treatment activates autocrine S1P/S1P2 signaling, which subsequently activates NFκB and leads to the proinflammatory responses in endothelial cells.


Subject(s)
Human Umbilical Vein Endothelial Cells/drug effects , Intercellular Adhesion Molecule-1/genetics , Lysophospholipids/metabolism , NF-kappa B/metabolism , Receptors, Lysosphingolipid/metabolism , Sphingosine/analogs & derivatives , Tumor Necrosis Factor-alpha/pharmacology , Vascular Cell Adhesion Molecule-1/genetics , Human Umbilical Vein Endothelial Cells/cytology , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Signal Transduction/drug effects , Sphingosine/metabolism , Sphingosine-1-Phosphate Receptors , Up-Regulation/drug effects
9.
Int J Endocrinol Metab ; 21(3): e136539, 2023 Jul.
Article in English | MEDLINE | ID: mdl-38028248

ABSTRACT

Background: It has been reported that sphingosine kinase (SK) 2 plays a role in maintaining metabolism and glucose homeostasis. However, the mechanism remains uncertain. Objectives: The present research aimed to further investigate the effect of SK2 knockout on high-fat diet (HFD)-induced obesity and metabolic regulation. Methods: Male SK2-/- and wild-type (WT) control mice were challenged with HFD for 8 weeks. Then, body composition, inguinal white adipose tissue (IWAT) histology, intraperitoneal glucose tolerance tests (IPGTT), and metabolic parameters were examined, and expression levels of uncoupling protein 1 (UCP1), a key molecular marker of thermogenesis, in IWAT were determined. Results: After 8 weeks of HFD challenge, compared with WT mice, SK2-/- mice displayed decreased whole body, epididymal white adipose tissue (EWAT) and IWAT weights, reduced fat/lean body mass ratios and inguinal adipocytes size; also, SK2-/- mice exhibited improved intraperitoneal glucose tolerance. Next, elevated energy expenditure was observed in SK2-/- mice compared with WT mice; however, neither food intake nor physical activity showed obvious difference between SK2-/- and WT mice. Furthermore, we found that the expressions of UCP1 was markedly increased in IWAT from SK2-/- mice. Conclusions: SK2-/- mice may resist HFD-induced obesity through increasing energy expenditure by promoting thermogenesis in the beige adipose tissue.

10.
J Biol Chem ; 286(39): 33832-40, 2011 Sep 30.
Article in English | MEDLINE | ID: mdl-21712392

ABSTRACT

Hydroxy fatty acids are critical lipid mediators involved in various pathophysiologic functions. We cloned and identified GPR31, a plasma membrane orphan G protein-coupled receptor that displays high affinity for the human 12-lipoxygenase-derived product 12-(S)-hydroxy-5,8,10,14-eicosatetraenoic acid (HETE). Thus, GPR31 is named 12-(S)-HETE receptor (12-HETER) in this study. The cloned 12-HETER demonstrated high affinity binding for 12-(S)-[(3)H]HETE (K(d) = 4.8 ± 0.12 nm). Also, 12-(S)-HETE efficiently and selectively stimulated GTPγS coupling in the membranes of 12-HETER-transfected cells (EC(50) = 0.28 ± 1.26 nm). Activating GTPγS coupling with 12-(S)-HETE proved to be both regio- and stereospecific. Also, 12-(S)-HETE/12-HETER interactions lead to activation of ERK1/2, MEK, and NFκB. Moreover, knocking down 12-HRTER specifically inhibited 12-(S)-HETE-stimulated cell invasion. Thus, 12-HETER represents the first identified high affinity receptor for the 12-(S)-HETE hydroxyl fatty acids.


Subject(s)
12-Hydroxy-5,8,10,14-eicosatetraenoic Acid/metabolism , Cell Membrane/metabolism , Receptors, G-Protein-Coupled/metabolism , 12-Hydroxy-5,8,10,14-eicosatetraenoic Acid/pharmacology , Animals , CHO Cells , COS Cells , Cell Membrane/genetics , Chlorocebus aethiops , Cloning, Molecular , Cricetinae , Cricetulus , Enzyme Activation/drug effects , Enzyme Activation/physiology , Guanosine 5'-O-(3-Thiotriphosphate)/pharmacology , Humans , MAP Kinase Kinase Kinases/genetics , MAP Kinase Kinase Kinases/metabolism , Mice , Mitogen-Activated Protein Kinase 1/genetics , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/genetics , Mitogen-Activated Protein Kinase 3/metabolism , NF-kappa B/genetics , NF-kappa B/metabolism , Protein Binding , Receptors, G-Protein-Coupled/genetics
11.
J Cell Physiol ; 219(3): 563-71, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19170074

ABSTRACT

It is well known that bone marrow-derived mesenchymal stem cells (MSCs) are involved in wound healing and regeneration responses. In this study, we globally profiled the proteome of MSCs to investigate critical factor(s) that may promote wound healing. Cysteine-rich protein 61 (Cyr61) was found to be abundantly present in MSCs. The presence of Cyr61 was confirmed by immunofluorescence staining and immunoblot analysis. Moreover, we showed that Cyr61 is present in the culture medium (secretome) of MSCs. The secretome of MSCs stimulates angiogenic response in vitro, and neovascularization in vivo. Depletion of Cyr61 completely abrogates the angiogenic-inducing capability of the MSC secretome. Importantly, addition of recombinant Cyr61 polypeptides restores the angiogenic activity of Cyr61-depleted secretome. Collectively, these data demonstrate that Cyr61 polypeptide in MSC secretome contributes to the angiogenesis-promoting activity, a key event needed for regeneration and repair of injured tissues. J. Cell. Physiol. 219: 563-571, 2009. (c) 2009 Wiley-Liss, Inc.


Subject(s)
Cysteine-Rich Protein 61/physiology , Mesenchymal Stem Cells/physiology , Neovascularization, Physiologic , Animals , Cells, Cultured , Collagen , Culture Media, Conditioned , Cysteine-Rich Protein 61/administration & dosage , Cysteine-Rich Protein 61/metabolism , Cysteine-Rich Protein 61/pharmacology , Drug Combinations , Endothelial Cells/cytology , Endothelial Cells/drug effects , Endothelial Cells/physiology , Humans , Laminin , Male , Mesenchymal Stem Cells/metabolism , Mice , Mice, Inbred C57BL , Mice, Nude , Neovascularization, Physiologic/drug effects , Proteoglycans , Recombinant Proteins/administration & dosage , Recombinant Proteins/pharmacology
12.
Histochem Cell Biol ; 131(2): 239-49, 2009 Feb.
Article in English | MEDLINE | ID: mdl-18936953

ABSTRACT

Sphingosine-1-phosphate (S1P) receptor subtype 1 (S1P(1)), a G-protein coupled receptor (GPCR), regulates many biological activities of endothelial cells (ECs). In this report, we show that S1P(1) receptors are present in the nuclei of ECs by using various biochemical and microscopic techniques such as cellular fractionation, immunogold labeling, and confocal microscopic analysis. Live cell imaging showed that plasma membrane S1P(1) receptors are rapidly internalized and subsequently translocated to nuclear compartment upon S1P stimulation. Utilizing membrane biotinylation technique further supports the notion that nuclear S1P(1) receptors were internalized from plasma membrane S1P(1) after ligand treatment. Moreover, nuclear S1P(1) is able to regulate the transcription of Cyr61 and CTGF, two growth factors functionally important in the regulation of vasculature. Collectively, these data suggest a novel S1P-S1P(1) signaling axis present in the nuclear compartment of endothelial cells, which may regulate biological responses of endothelium.


Subject(s)
Active Transport, Cell Nucleus , Connective Tissue Growth Factor/genetics , Cysteine-Rich Protein 61/genetics , Endothelial Cells/metabolism , Receptors, Lysosphingolipid/metabolism , Transcription, Genetic , Cell Membrane , Endothelium, Vascular , Humans , Ligands , Lysophospholipids/metabolism , Sphingosine/analogs & derivatives , Sphingosine/metabolism
13.
J Cell Biochem ; 104(5): 1793-802, 2008 Aug 01.
Article in English | MEDLINE | ID: mdl-18348263

ABSTRACT

Conditioned medium (secretome) derived from an enriched stem cell culture stimulates chemotaxis of human fibroblasts. These cells are classified as multipotent murine mesenchymal stromal cells (mMSC) by immunochemical analysis of marker proteins. Proteomic analysis of mMSC secretome identifies nineteen secreted proteins, including extracellular matrix structural proteins, collagen processing enzymes, pigment epithelium-derived factor (PEDF) and cystatin C. Immunodepletion and reconstitution experiments show that PEDF is the predominant fibroblast chemoattractant in the conditioned medium, and immunofluorescence microscopy shows strong staining for PEDF in the cytoplasm, at the cell surface, and in intercellular space between mMSCs. This stimulatory effect of PEDF on fibroblast chemotaxis is in contrast to the PEDF-mediated inhibition of endothelial cell migration, reported previously. These differential functional effects of PEDF toward fibroblasts and endothelial cells may serve to program an ordered temporal sequence of scaffold building followed by angiogenesis during wound healing.


Subject(s)
Chemotaxis , Eye Proteins/metabolism , Fibroblasts/cytology , Mesenchymal Stem Cells/metabolism , Nerve Growth Factors/metabolism , Serpins/metabolism , Amino Acid Sequence , Animals , Cattle , Cell Line , Culture Media, Conditioned , Eye Proteins/chemistry , Humans , Mass Spectrometry , Mice , Molecular Sequence Data , Nerve Growth Factors/chemistry , Proteomics , Reproducibility of Results , Serpins/chemistry
14.
J Atheroscler Thromb ; 10(3): 125-31, 2003.
Article in English | MEDLINE | ID: mdl-14564080

ABSTRACT

The signaling and functions of the Endothelial Differentiation Gene (EDG) family of G protein-coupled receptors have been extensively elucidated. All the members of EDG family were shown to be receptors for lysosphingolipids or lysophospholipids. EDG-1, the prototype of EDG family receptors, is a high affinity receptor for serum-borne bioactive lipid, Sphingosine-1-phosphate (S1P). S1P, secreted by thrombotic platelets, has been shown to regulate a variety of cellular responses, including survival, cytoskeletal remodeling, chemotaxis etc, via the activation of cell surface EDG receptors. Recently, a novel function of S1P in modulating angiogenic response has been demonstrated. This review will focus on S1P/ EDG-1 signaling in endothelial activation, in particular in the S1P-mediated adherens junctions assembly and chemotaxis in endothelial cells.


Subject(s)
Endothelium, Vascular/physiology , Lysophospholipids , Platelet Activation/physiology , Signal Transduction/physiology , Sphingosine/analogs & derivatives , Sphingosine/physiology , Humans , Immediate-Early Proteins/physiology , Neovascularization, Physiologic/physiology , Receptors, G-Protein-Coupled/physiology , Receptors, Lysophospholipid
15.
J Biochem Pharmacol Res ; 1(4): 197-203, 2013 Dec.
Article in English | MEDLINE | ID: mdl-25383272

ABSTRACT

Sphingosine-1-phosphate (S1P), a serum-borne bioactive lipid, regulates various physiological functions. We observed that the S1P receptor subtype 1 (S1P1), a high affinity G-protein coupled receptor of S1P, is the major S1P receptor expressed in the Kit+/Sca-1+/Lin- (KSL) hematopoietic stem progenitor cells (HSPCs, KSL-HSPCs). In this study, we investigate function of S1P1 receptors in the regulation of HSPC mobilization in animals. Treatment with SEW2871, a specific agonist of S1P1, had no effect on KSL-HSPC mobilization. In addition, mice pretreated with SEW2871 followed by AMD3100, a well-known activator of KSL-HSPC mobilization by antagonizing the stromal-derived factor-1 (SDF-1)/C-X-C chemokine receptor type 4 (CXCR4) signaling axis, did not enhance the AMD3100-induced KSL-HSPC mobilization. In contrast, pretreatment of (R)-3-amino-4-(3-hexylphenylamino)-4-oxobutyl phosphonic acid (W146), a selective antagonist of S1P1, significantly augments AMD3100-induced KSL-HSPC mobilization into peripheral blood. The inactive enantiomer W140 was incapable of enhancing the AMD3100-induced KSL-HSPC mobilization. Moreover, treatment with selective antagonists for S1P2 and S1P3 had no effects on AMD3100-mediated KSL-HSPC mobilization. Collectively, our data suggest that S1P/S1P1 signaling regulates the SDF-1/CXCR4-mediated retention of KSL-HSPCs in bone marrow microenvironment.

16.
Int J Oncol ; 40(5): 1619-26, 2012 May.
Article in English | MEDLINE | ID: mdl-22344462

ABSTRACT

Sphingosine-1-phosphate (S1P) regulates a wide array of biological functions. However, the role of S1P signaling in tumorigenesis remains to be elucidated. In this study, we show that S1P receptor subtype 3 (S1P3) is markedly up-regulated in a subset of lung adenocarcinoma cells compared to normal lung epithelial cells. Specific knockdown of S1P3 receptors inhibits proliferation and anchorage-independent growth of lung adenocarcinoma cells. Mechanistically, we demonstrate that S1P3 signaling increases epidermal growth factor receptor (EGFR) expression via the Rho kinase (ROCK) pathway in lung adenocarcinoma cells. Nuclear run-off analysis indicates that S1P/S1P3 signaling transcriptionally increases EGFR expression. Knockdown of S1P3 receptors diminishes the S1P-stimulated EGFR expression in lung adenocarcinoma cells. Moreover, S1P treatment greatly enhances EGF-stimulated colony formation, proliferation and invasion of lung adenocarcinoma cells. Together, these results suggest that the enhanced S1P3-EGFR signaling axis may contribute to the tumorigenesis or progression of lung adenocarcinomas.


Subject(s)
Adenocarcinoma/metabolism , Carcinoma, Lewis Lung/metabolism , Cell Movement , Cell Proliferation , ErbB Receptors/metabolism , Lung Neoplasms/metabolism , Receptors, Lysosphingolipid/metabolism , Signal Transduction , Adenocarcinoma/genetics , Adenocarcinoma of Lung , Animals , Carcinoma, Lewis Lung/genetics , Cell Line, Tumor , Epidermal Growth Factor/metabolism , ErbB Receptors/genetics , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Humans , Lung Neoplasms/genetics , Lysophospholipids/metabolism , Mice , Neoplasm Invasiveness , RNA Interference , RNA, Messenger/metabolism , Receptors, Lysosphingolipid/genetics , Sphingosine/analogs & derivatives , Sphingosine/metabolism , Sphingosine-1-Phosphate Receptors , Time Factors , Transcriptional Activation , Transfection , Up-Regulation , rho-Associated Kinases/metabolism
17.
World J Biol Chem ; 2(1): 1-13, 2011 Jan 26.
Article in English | MEDLINE | ID: mdl-21472036

ABSTRACT

Most hematopoietic stem progenitor cells (HSPCs) reside in bone marrow (BM), but a small amount of HSPCs have been found to circulate between BM and tissues through blood and lymph. Several lines of evidence suggest that sphingosine-1-phosphate (S1P) gradient triggers HSPC egression to blood circulation after mobilization from BM stem cell niches. Stem cells also visit certain tissues. After a temporary 36 h short stay in local tissues, HSPCs go to lymph in response to S1P gradient between lymph and tissue and eventually enter the blood circulation. S1P also has a role in the guidance of the primitive HSPCs homing to BM in vivo, as S1P analogue FTY720 treatment can improve HSPC BM homing and engraftment. In stress conditions, various stem cells or progenitor cells can be attracted to local injured tissues and participate in local tissue cell differentiation and tissue rebuilding through modulation the expression level of S1P(1), S1P(2) or S1P(3) receptors. Hence, S1P is important for stem cells circulation in blood system to accomplish its role in body surveillance and injury recovery.

18.
Am J Physiol Heart Circ Physiol ; 296(1): H33-42, 2009 Jan.
Article in English | MEDLINE | ID: mdl-19011048

ABSTRACT

Sphingosine-1-phosphate (S1P) regulates various molecular and cellular events in cultured endothelial cells, such as cytoskeletal restructuring, cell-extracellular matrix interactions, and intercellular junction interactions. We utilized the venular leakage model of the cremaster muscle vascular bed in Sprague-Dawley rats to investigate the role of S1P signaling in regulation of microvascular permeability. S1P signaling is mediated by the S1P family of G protein-coupled receptors (S1P(1-5) receptors). S1P(1) and S1P(2) receptors, which transduce stimulatory and inhibitory signaling, respectively, are expressed in the endothelium of the cremaster muscle vasculature. S1P administration alone via the carotid artery was unable to protect against histamine-induced venular leakage of the cremaster muscle vascular bed in Sprague-Dawley rats. However, activation of S1P(1)-mediated signaling by SEW2871 and FTY720, two agonists of S1P(1), significantly inhibited histamine-induced microvascular leakage. Treatment with VPC 23019 to antagonize S1P(1)-regulated signaling greatly potentiated histamine-induced venular leakage. After inhibition of S1P(2) signaling by JTE-013, a specific antagonist of S1P(2), S1P was able to protect microvascular permeability in vivo. Moreover, endothelial tight junctions and barrier function were regulated by S1P(1)- and S1P(2)-mediated signaling in a concerted manner in cultured endothelial cells. These data suggest that the balance between S1P(1) and S1P(2) signaling regulates the homeostasis of microvascular permeability in the peripheral circulation and, thus, may affect total peripheral vascular resistance.


Subject(s)
Capillary Permeability/physiology , Lysophospholipids/physiology , Muscle, Skeletal/blood supply , Receptors, Lysosphingolipid/physiology , Signal Transduction/physiology , Sphingosine/analogs & derivatives , Aging/physiology , Animals , Carotid Arteries/physiology , Fluorescent Antibody Technique , Histamine/pharmacology , Histamine Agonists/pharmacology , Histamine Antagonists/pharmacology , Homeostasis , Infusions, Intra-Arterial , Lysophospholipids/administration & dosage , Lysophospholipids/pharmacology , Male , Muscle, Skeletal/physiology , Rats , Rats, Sprague-Dawley , Regional Blood Flow/physiology , Sphingosine/administration & dosage , Sphingosine/pharmacology , Sphingosine/physiology
19.
Histochem Cell Biol ; 129(5): 579-88, 2008 May.
Article in English | MEDLINE | ID: mdl-18247041

ABSTRACT

Integrins, a family of transmembrane heterodimeric polypeptides, mediate various biological responses including cell adhesion and migration. In this report, we show that sphingosine-1-phosphate (S1P) activates integrin alpha v beta 3 in endothelial cells (ECs) via the sphingosine-1-phosphate receptor subtype 1 (S1P1)-mediated signaling pathway. S1P treatment results in the activation of integrin alpha v beta 3 in the lamellipodia region of ECs, suggesting that integrin alpha v beta 3 plays a critical role in the S1P-stimulated chemotactic response of ECs. Indeed, S1P treatment induces the association of focal adhesion kinase (FAK) and cytoskeletal proteins with integrin alpha v beta 3, the ligation of alpha v and beta 3 subunits, as well as enhances endothelial migration on vitronectin-coated substrata. Knockdown endothelial S1P1 receptor, treatments with pertussis toxin or dominant-negative-Rho family GTPases abrogates the S1P-induced integrin alpha v beta 3 activation in ECs. Consequently, these treatments markedly inhibit the S1P-induced endothelial migratory response on vitronectin-coated substrata. Collectively, these data indicate that the S1P-mediated signaling via the S1P1/Gi/Rho GTPases pathway activates integrin alpha v beta 3, which is indispensable for S1P-stimulated chemotactic response of ECs.


Subject(s)
Chemotaxis/drug effects , Endothelial Cells/drug effects , Integrin alphaVbeta3/metabolism , Lysophospholipids/pharmacology , Sphingosine/analogs & derivatives , rho GTP-Binding Proteins/metabolism , Cell Adhesion/drug effects , Cell Line , Cell Movement/drug effects , Chemotaxis/physiology , Cytoskeletal Proteins/metabolism , Endothelial Cells/cytology , Endothelial Cells/metabolism , Fluorescent Antibody Technique , Focal Adhesion Protein-Tyrosine Kinases/metabolism , Gene Silencing , Humans , Immunoblotting , Pertussis Toxin/pharmacology , RNA, Small Interfering/genetics , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Receptors, Lysosphingolipid/genetics , Receptors, Lysosphingolipid/metabolism , Sphingosine/pharmacology , Vitronectin/metabolism , rho GTP-Binding Proteins/genetics
20.
J Immunol ; 180(3): 1818-25, 2008 Feb 01.
Article in English | MEDLINE | ID: mdl-18209079

ABSTRACT

IFN-beta production is a critical step in human innate immune responses and is primarily controlled at the transcription level by highly ordered mechanisms. IFN-beta can be induced by pattern-recognition receptors such as the TLR4. S1P1 is a G protein-coupled receptor, which has a high affinity for sphingosine 1-phosphate (S1P). Although many of the receptors and signaling pathways leading to the expression of IFN-beta have been identified and characterized, it is still unclear how IFN-beta is regulated in primary human gingival epithelial cells (HGECs). In this study, we demonstrate that S1P1 and TLR4, acting in unison, play an important role in IFN-beta expression at the protein and mRNA level in HGECs. We demonstrate that the expression of both IFN-beta and IFN-inducible protein-10 (CXCL-10) is significantly up-regulated by LPS and S1P or LPS and a specific S1P1 agonist. This enhanced innate immune response is attenuated in HGECs by small interfering RNA knockdown of either TLR4 or S1P1. Moreover, we show that triggering of TLR4 results in the increased expression of S1P1 receptors. Furthermore, we found that IFN-regulatory factor 3 activation was maximized by LPS and S1P through PI3K. Our data show that triggering TLR4 increases S1P1, such that both TLR4 and S1P1 acting through PI3K enhancement of IFN-regulatory factor 3 activation increase IFN-beta expression in epithelial cells. The functional association between TLR4 and the S1P1 receptor demonstrates a novel mechanism in the regulation of IFN-beta and CXCL-10 in human primary gingival epithelial cells.


Subject(s)
Gingiva/immunology , Interferon-beta/metabolism , Lysophospholipids/physiology , Sphingosine/analogs & derivatives , Toll-Like Receptor 4/physiology , Cells, Cultured , Chemokine CXCL10/metabolism , Epithelial Cells/drug effects , Epithelial Cells/immunology , Gingiva/cytology , Gingiva/drug effects , Humans , Interferon Regulatory Factor-3/metabolism , Interferon-beta/genetics , Lipopolysaccharides/pharmacology , Lysophospholipids/pharmacology , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation , Receptors, Lysosphingolipid/agonists , Receptors, Lysosphingolipid/metabolism , Sphingosine/pharmacology , Sphingosine/physiology
SELECTION OF CITATIONS
SEARCH DETAIL