1
|
Feng Y, Mischler WJ, Gurung AC, Kavanagh TR, Androsov G, Sadow PM, Herbert ZT, Priolo C. Therapeutic Targeting of the Secreted Lysophospholipase D Autotaxin Suppresses Tuberous Sclerosis Complex-Associated Tumorigenesis. Cancer Res 2020; 80:2751-2763. [PMID: 32393662 DOI: 10.1158/0008-5472.can-19-2884] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Revised: 03/25/2020] [Accepted: 05/05/2020] [Indexed: 12/14/2022]
Abstract
Tuberous sclerosis complex (TSC) is an autosomal dominant disease characterized by multiorgan hamartomas, including renal angiomyolipomas and pulmonary lymphangioleiomyomatosis (LAM). TSC2 deficiency leads to hyperactivation of mTOR Complex 1 (mTORC1), a master regulator of cell growth and metabolism. Phospholipid metabolism is dysregulated upon TSC2 loss, causing enhanced production of lysophosphatidylcholine (LPC) species by TSC2-deficient tumor cells. LPC is the major substrate of the secreted lysophospholipase D autotaxin (ATX), which generates two bioactive lipids, lysophosphatidic acid (LPA) and sphingosine-1-phosphate (S1P). We report here that ATX expression is upregulated in human renal angiomyolipoma-derived TSC2-deficient cells compared with TSC2 add-back cells. Inhibition of ATX via the clinically developed compound GLPG1690 suppressed TSC2-loss associated oncogenicity in vitro and in vivo and induced apoptosis in TSC2-deficient cells. GLPG1690 suppressed AKT and ERK1/2 signaling and profoundly impacted the transcriptome of these cells while inducing minor gene expression changes in TSC2 add-back cells. RNA-sequencing studies revealed transcriptomic signatures of LPA and S1P, suggesting an LPA/S1P-mediated reprogramming of the TSC lipidome. In addition, supplementation of LPA or S1P rescued proliferation and viability, neutral lipid content, and AKT or ERK1/2 signaling in human TSC2-deficient cells treated with GLPG1690. Importantly, TSC-associated renal angiomyolipomas have higher expression of LPA receptor 1 and S1P receptor 3 compared with normal kidney. These studies increase our understanding of TSC2-deficient cell metabolism, leading to novel potential therapeutic opportunities for TSC and LAM. SIGNIFICANCE: This study identifies activation of the ATX-LPA/S1P pathway as a novel mode of metabolic dysregulation upon TSC2 loss, highlighting critical roles for ATX in TSC2-deficient cell fitness and in TSC tumorigenesis.
Collapse
Affiliation(s)
- You Feng
- Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts
- Harvard Medical School, Boston, Massachusetts
| | - William J Mischler
- Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts
- Harvard Medical School, Boston, Massachusetts
| | - Ashish C Gurung
- Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts
- Harvard Medical School, Boston, Massachusetts
| | - Taylor R Kavanagh
- Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts
- Harvard Medical School, Boston, Massachusetts
| | - Grigoriy Androsov
- Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts
- Harvard Medical School, Boston, Massachusetts
| | - Peter M Sadow
- Harvard Medical School, Boston, Massachusetts
- Department of Pathology, Massachusetts General Hospital, Boston, Massachusetts
| | - Zachary T Herbert
- Harvard Medical School, Boston, Massachusetts
- Molecular Biology Core Facilities, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Carmen Priolo
- Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts.
- Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
2
|
Alphastatin-Loaded Chitosan Nanoparticle Preparation and Its Antiangiogenic Effect on Lung Carcinoma. INT J POLYM SCI 2019. [DOI: 10.1155/2019/2751384] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Alphastatin is a 24-amino acid peptide and can suppress tumor angiogenesis by inhibiting both the migration and tubule formation of vascular endothelial cells. However, the anticancer effect of Alphastatin is limited due to the short half-life and degradation in the body. In this study, Alphastatin-loaded chitosan nanoparticles (AsCs NPs) were prepared with an initial concentration of 2 mg/ml for chitosan and 1 mg/ml for Alphastatin. AsCs NPs presented the encapsulation efficiency of 32.4%, the mean particle size of 387.4 nm, the polydispersity index of 0.223, and the zeta potential of +28.1 mV. AsCs NPs have a sustained release for 6 days and were stable in serum for at least 24 hours. And the NPs could preserve the integrity of encapsulated Alphastatin and released Alphastatin for 24 hours. In a subcutaneous LA975 lung carcinoma xenograft T739 mouse model, AsCs NPs significantly inhibited the tumor growth, tumor volume, and microvessel density (MVD), and the antitumor effect was even stronger than that of Alphastatin. In addition, the VEGF-induced tube formation of HUVEC could be inhibited by AsCs NPs in vitro and the serum containing AsCs NPs, and the protein level of SphK1 in HUVEC was also decreased by AsCs NPs, suggesting an inhibitory effect of AsCs NPs on the SphK1-S1P signaling pathway. Furthermore, hemolysis assay showed a safety on blood compatibility of AsCs NPs. Our study indicated that AsCs NPs inhibited the SphK1-S1P signaling pathway and enhanced the antiangiogenic effect of Alphastatin both in vitro and in vivo.
Collapse
|
3
|
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.
Collapse
Affiliation(s)
- Yu Hisano
- Vascular Biology Program, Boston Children's Hospital, Department of Surgery, Harvard Medical School, 300 Longwood Avenue, Boston, MA 02115, United States
| | - Timothy Hla
- Vascular Biology Program, Boston Children's Hospital, Department of Surgery, Harvard Medical School, 300 Longwood Avenue, Boston, MA 02115, United States.
| |
Collapse
|
4
|
Vrzalikova K, Ibrahim M, Vockerodt M, Perry T, Margielewska S, Lupino L, Nagy E, Soilleux E, Liebelt D, Hollows R, Last A, Reynolds G, Abdullah M, Curley H, Care M, Krappmann D, Tooze R, Allegood J, Spiegel S, Wei W, Woodman CBJ, Murray PG. S1PR1 drives a feedforward signalling loop to regulate BATF3 and the transcriptional programme of Hodgkin lymphoma cells. Leukemia 2018; 32:214-223. [PMID: 28878352 PMCID: PMC5737877 DOI: 10.1038/leu.2017.275] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2017] [Revised: 08/09/2017] [Accepted: 08/14/2017] [Indexed: 12/15/2022]
Abstract
The Hodgkin/Reed-Sternberg cells of classical Hodgkin lymphoma (HL) are characterised by the aberrant activation of multiple signalling pathways. Here we show that a subset of HL displays altered expression of sphingosine-1-phosphate (S1P) receptors (S1PR)s. S1P activates phosphatidylinositide 3-kinase (PI3-K) in these cells that is mediated by the increased expression of S1PR1 and the decreased expression of S1PR2. We also showed that genes regulated by the PI3-K signalling pathway in HL cell lines significantly overlap with the transcriptional programme of primary HRS cells. Genes upregulated by the PI3-K pathway included the basic leucine zipper transcription factor, ATF-like 3 (BATF3), which is normally associated with the development of dendritic cells. Immunohistochemistry confirmed that BATF3 was expressed in HRS cells of most HL cases. In contrast, in normal lymphoid tissues, BATF3 expression was confined to a small fraction of CD30-positive immunoblasts. Knockdown of BATF3 in HL cell lines revealed that BATF3 contributed to the transcriptional programme of primary HRS cells, including the upregulation of S1PR1. Our data suggest that disruption of this potentially oncogenic feedforward S1P signalling loop could provide novel therapeutic opportunities for patients with HL.
Collapse
Affiliation(s)
- K Vrzalikova
- Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, UK
| | - M Ibrahim
- Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, UK
| | - M Vockerodt
- Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, UK
- Institute of Anatomy and Cell Biology, Georg-August University of Göttingen, Göttingen, Germany
| | - T Perry
- Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, UK
| | - S Margielewska
- Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, UK
| | - L Lupino
- Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, UK
| | - E Nagy
- Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, UK
| | - E Soilleux
- Department of Cellular Pathology, John Radcliffe Hospital, Oxford, UK
| | - D Liebelt
- Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, UK
| | - R Hollows
- Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, UK
| | - A Last
- Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, UK
| | - G Reynolds
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK
| | - M Abdullah
- Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, UK
- Department of Pathology, Universiti Putra Malaysia, Selangor, Malaysia
| | - H Curley
- Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, UK
| | - M Care
- Leeds Institute of Cancer and Pathology, University of Leeds, Leeds, UK
| | - D Krappmann
- Research Unit Cellular Signal Integration, Helmholtz Zentrum München, Neuherberg, Germany
| | - R Tooze
- Leeds Institute of Cancer and Pathology, University of Leeds, Leeds, UK
| | - J Allegood
- Department of Biochemistry and Molecular Biology and Massey Cancer Center, Virginia Commonwealth University School of Medicine, Richmond, VA, USA
| | - S Spiegel
- Department of Biochemistry and Molecular Biology and Massey Cancer Center, Virginia Commonwealth University School of Medicine, Richmond, VA, USA
| | - W Wei
- Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, UK
- Sheffield Institute of Translational Neuroscience, University of Sheffield, Sheffield, UK
| | - C B J Woodman
- Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, UK
| | - P G Murray
- Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, UK
- Department of Clinical and Molecular Pathology, Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University, Olomouc, Czech Republic
| |
Collapse
|
5
|
Cho JH, Han JS. Phospholipase D and Its Essential Role in Cancer. Mol Cells 2017; 40:805-813. [PMID: 29145720 PMCID: PMC5712509 DOI: 10.14348/molcells.2017.0241] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2017] [Revised: 10/16/2017] [Accepted: 11/11/2017] [Indexed: 11/27/2022] Open
Abstract
The role of phospholipase D (PLD) in cancer development and management has been a major area of interest for researchers. The purpose of this mini-review is to explore PLD and its distinct role during chemotherapy including anti-apoptotic function. PLD is an enzyme that belongs to the phospholipase super family and is found in a broad range of organisms such as viruses, yeast, bacteria, animals, and plants. The function and activity of PLD are widely dependent on and regulated by neurotransmitters, hormones, small monomeric GTPases, and lipids. A growing body of research has shown that PLD activity is significantly increased in cancer tissues and cells, indicating that it plays a critical role in signal transduction, cell proliferation, and anti-apoptotic processes. In addition, recent studies show that PLD is a downstream transcriptional target of proteins that contribute to inflammation and carcinogenesis such as Sp1, NFκB, TCF4, ATF-2, NFATc2, and EWS-Fli. Thus, compounds that inhibit expression or activity of PLD in cells can be potentially useful in reducing inflammation and sensitizing resistant cancers during chemotherapy.
Collapse
Affiliation(s)
- Ju Hwan Cho
- Arthur G. James Cancer Hospital Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio 4321,
USA
| | - Joong-Soo Han
- Biomedical Research Institute and Department of Biochemistry & Molecular Biology, College of Medicine, Hanyang University, Seoul 04763,
Korea
| |
Collapse
|
6
|
Pi J, Wu X, Yang S, Zeng P, Feng Y. Rapid identification of erythrocyte phospholipids in Sprague-Dawley rats by ultra high performance liquid chromatography with electrospray ionization quadrupole time-of-flight tandem mass spectrometry. J Sep Sci 2015; 38:886-93. [PMID: 25564825 DOI: 10.1002/jssc.201401120] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2014] [Revised: 11/24/2014] [Accepted: 12/22/2014] [Indexed: 12/13/2022]
Abstract
A rapid, sensitive, and reliable approach for analyzing five kinds of erythrocyte phospholipids in Sprague-Dawley rats was provided by ultra high performance liquid chromatography coupled with electrospray ionization quadrupole time-of-flight tandem mass spectrometry with MassLynx(TM) MassFragment. Improving conventional high performance liquid chromatography techniques, ultra high performance liquid chromatography integrated with quadrupole time-of-flight tandem mass spectrometry offers high sensitivity and increased analytical speed by using columns packed with sub-2 μm particles (1.7 μm), which allows a faster separation to be achieved. Through this method, 83 phospholipids were tentatively characterized based on their mass spectra and tandem mass spectra, as well as by matching the in-house formula database within a mass error of 5 ppm, including 40 phosphatidylcholines, 24 phosphatidyl ethanolamines, three phosphatidylinositols, six phosphatidylserines, and ten sphingomyelins. Our present results proved that the established method could be used to qualitatively analyze complex erythrocyte phospholipids in Sprague-Dawley rats and provide a useful data base for pharmacology and phospholipidomics to seek potential biomarkers of disease prediction.
Collapse
Affiliation(s)
- Juanjuan Pi
- Central laboratory, Guangdong Pharmaceutical University, Guangzhou, P. R. China
| | | | | | | | | |
Collapse
|
7
|
Zhang L, Wang X, Bullock AJ, Callea M, Shah H, Song J, Moreno K, Visentin B, Deutschman D, Alsop DC, Atkins MB, Mier JW, Signoretti S, Bhasin M, Sabbadini RA, Bhatt RS. Anti-S1P Antibody as a Novel Therapeutic Strategy for VEGFR TKI-Resistant Renal Cancer. Clin Cancer Res 2015; 21:1925-1934. [PMID: 25589614 DOI: 10.1158/1078-0432.ccr-14-2031] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2014] [Accepted: 12/13/2014] [Indexed: 01/22/2023]
Abstract
PURPOSE VEGFR2 tyrosine kinase inhibition (TKI) is a valuable treatment approach for patients with metastatic renal cell carcinoma (RCC). However, resistance to treatment is inevitable. Identification of novel targets could lead to better treatment for patients with TKI-naïve or -resistant RCC. EXPERIMENTAL DESIGN In this study, we performed transcriptome analysis of VEGFR TKI-resistant tumors in a murine model and discovered that the SPHK-S1P pathway is upregulated at the time of resistance. We tested sphingosine-1-phosphate (S1P) pathway inhibition using an anti-S1P mAb (sphingomab), in two mouse xenograft models of RCC, and assessed tumor SPHK expression and S1P plasma levels in patients with metastatic RCC. RESULTS Resistant tumors expressed several hypoxia-regulated genes. The SPHK1 pathway was among the most highly upregulated pathways that accompanied resistance to VEGFR TKI therapy. SPHK1 was expressed in human RCC, and the product of SPHK1 activity, S1P, was elevated in patients with metastatic RCC, suggesting that human RCC behavior could, in part, be due to overproduction of S1P. Sphingomab neutralization of extracellular S1P slowed tumor growth in both mouse models. Mice bearing tumors that had developed resistance to sunitinib treatment also exhibited tumor growth suppression with sphingomab. Sphingomab treatment led to a reduction in tumor blood flow as measured by MRI. CONCLUSIONS Our findings suggest that S1P inhibition may be a novel therapeutic strategy in patients with treatment-naïve RCC and also in the setting of resistance to VEGFR TKI therapy.
Collapse
Affiliation(s)
- Liang Zhang
- Division of Hematology-Oncology and Cancer Biology, Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Avenue, Boston, Massachusetts, 02215, United States of America
| | - Xiaoen Wang
- Division of Hematology-Oncology and Cancer Biology, Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Avenue, Boston, Massachusetts, 02215, United States of America.,Department of Radiology, Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Avenue, Boston, Massachusetts, 02215, United States of America
| | - Andrea J Bullock
- Division of Hematology-Oncology and Cancer Biology, Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Avenue, Boston, Massachusetts, 02215, United States of America
| | - Marcella Callea
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, 75 Francis Street, Boston, Massachusetts, 02115, United States of America
| | - Harleen Shah
- Division of Interdisciplinary Medicine and Biotechnology, and Genomics and Proteomics Center, Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Avenue, Boston, Massachusetts, 02215, United States of America
| | - Jiaxi Song
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, 75 Francis Street, Boston, Massachusetts, 02115, United States of America
| | - Kelli Moreno
- Lpath Inc., 4025 Sorrento Valley Blvd. San Diego, CA, 92121, United States of America
| | - Barbara Visentin
- Lpath Inc., 4025 Sorrento Valley Blvd. San Diego, CA, 92121, United States of America
| | - Douglas Deutschman
- Department of Biology, San Diego State University, 5500 Campanile Dr. San Diego, CA. 92182-4614, United States of America
| | - David C Alsop
- Department of Radiology, Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Avenue, Boston, Massachusetts, 02215, United States of America
| | - Michael B Atkins
- Departments of Oncology and Medicine, Georgetown-Lombardi Comprehensive Cancer Center, 3970 Reservoir Road, NW, Washington, DC. United States of America
| | - James W Mier
- Division of Hematology-Oncology and Cancer Biology, Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Avenue, Boston, Massachusetts, 02215, United States of America
| | - Sabina Signoretti
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, 75 Francis Street, Boston, Massachusetts, 02115, United States of America
| | - Manoj Bhasin
- Division of Interdisciplinary Medicine and Biotechnology, and Genomics and Proteomics Center, Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Avenue, Boston, Massachusetts, 02215, United States of America
| | - Roger A Sabbadini
- Lpath Inc., 4025 Sorrento Valley Blvd. San Diego, CA, 92121, United States of America
| | - Rupal S Bhatt
- Division of Hematology-Oncology and Cancer Biology, Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Avenue, Boston, Massachusetts, 02215, United States of America
| |
Collapse
|
8
|
Egom EE, Rose RA, Neyses L, Soran H, Cleland JGF, Mamas MA. Activation of sphingosine-1-phosphate signalling as a potential underlying mechanism of the pleiotropic effects of statin therapy. Crit Rev Clin Lab Sci 2013; 50:79-89. [DOI: 10.3109/10408363.2013.813013] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
9
|
Zhao CG, Qin J, He XJ, Guan YC, Jia Y, Lei W. Sphingosine-1-phosphate is a possible fibrogenic factor in gluteal muscle fibrosis. Physiol Res 2013; 62:691-9. [PMID: 23869887 DOI: 10.33549/physiolres.932441] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Gluteal muscle contracture (GMC) is a chronic fibrotic disease of gluteal muscles due to multiple etiologies. The main pathologic process is characterized by proliferation of fibroblasts and excessive accumulation of collagen in the extracellular matrix of the muscle. Sphingosine-1-phosphate (S1P) is a bioactive sphingolipid and has been reported to be associated with various fibrotic diseases. However, the role of S1P in GMC remains unknown. Here in this article, High-performance liquid chromatography and immunohistochemistry were applied to evaluate S1P localization and expression in clinical samples from patients with GMC, Quantitative real time PCR, Western blot, and enzyme-linked immunosorbent assay were used to explore the link between transforming growth factor-beta1 (TGF-beta1), plasminogen activator inhibitor-1 (PAI-1) and S1P. The results showed that S1P was enhanced in contraction band (CB) tissues. Studies using the cell proliferation and transformation assay indicated that exogenous S1P stimulated CB fibroblast proliferation in a time-dependent manner and in higher concentration also in a dose-dependent manner. Furthermore, we demonstrated that S1P not only promoted collagen type I production, but also up-regulated mRNA and protein expression of transforming growth factor-beta1 and plasminogen activator inhibitor-1. These findings suggest that S1P may regulate increased synthesis of collagen and other fibrogenic factors, and significantly contributes to the process of gluteal muscle scarring in patients with GMC.
Collapse
Affiliation(s)
- C G Zhao
- Orthopedic Center of Chinese PLA, Urumqi General Hospital of Lanzhou Military Region, Urumqi, Xinjiang, China.
| | | | | | | | | | | |
Collapse
|
10
|
Gomez-Muñoz A, Gangoiti P, Arana L, Ouro A, Rivera IG, Ordoñez M, Trueba M. New insights on the role of ceramide 1-phosphate in inflammation. Biochim Biophys Acta Mol Cell Biol Lipids 2013; 1831:1060-6. [DOI: 10.1016/j.bbalip.2013.02.001] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2012] [Revised: 01/29/2013] [Accepted: 02/05/2013] [Indexed: 01/08/2023]
|
11
|
Lee H, Park HS, Hong SH, Choi OK, Cho SD, Park J, Oh JE, Chung SS, Jung HS, Park KS. 4-Deoxypyridoxine improves the viability of isolated pancreatic islets ex vivo. Islets 2013; 5:116-21. [PMID: 23756681 DOI: 10.4161/isl.25254] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
The successful islet transplantation, for the treatment of type 1 diabetes, depends on the quantity and the quality of transplanted islets. Previously, it has reported that the significant loss of isolated islet mass could be prevented by sphingolipid metabolite, sphinogosine 1-phophate (S1P). This study was performed to elucidate whether the beneficial effects of S1P maintaining isolated pancreatic islets ex vivo are mimicked by modulation of intracellular S1P. We tested the in vitro effect of various agents that modulate intracellular S1P levels in insulinoma cell lines and isolated islets to compare their anti-apoptotic effects with that of S1P. As results, we discovered that 4-deoxypyridoxine (DOP), which inhibits the degradation of intracellular S1P by inhibiting S1P lyase (SPL) activity, minimized the chemically induced apoptosis of insulinoma cell lines as S1P did. Also, supplementation of DOP in the culture media protected the regression of isolated islets that have been maintained ex vivo at least for 18 h providing the evidence of increasing viability of isolated islets with DOP, which impaired SPL activity. In conclusion, these results suggest that the application of SPL inhibitors could be considered as a supplement for the maintenance of viable islets isolated from donor sources in the process of islet transplantation.
Collapse
Affiliation(s)
- Hakmo Lee
- Department of Internal Medicine; Seoul National University College of Medicine; Seoul, Republic of Korea; Innovative Research Institute for Cell Therapy; Seoul National University Hospital; Seoul, Republic of Korea; Biomedical Research Institute; Seoul National University Hospital; Seoul, Republic of Korea
| | | | | | | | | | | | | | | | | | | |
Collapse
|
12
|
Kim YD, Han KT, Lee J, Park CG, Kim MY, Shahi PK, Zuo DC, Choi S, Jun JY. Effects of sphingosine-1-phosphate on pacemaker activity of interstitial cells of Cajal from mouse small intestine. Mol Cells 2013; 35:79-86. [PMID: 23307289 PMCID: PMC3887851 DOI: 10.1007/s10059-013-2282-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2012] [Revised: 12/03/2012] [Accepted: 12/03/2012] [Indexed: 01/28/2023] Open
Abstract
Interstitial cells of Cajal (ICC) are the pacemaker cells that generate the rhythmic oscillation responsible for the production of slow waves in gastrointestinal smooth muscle. Spingolipids are known to present in digestive system and are responsible for multiple important physiological and pathological processes. In this study, we are interested in the action of sphingosine 1-phosphate (S1P) on ICC. S1P depolarized the membrane and increased tonic inward pacemaker currents. FTY720 phosphate (FTY720P, an S1P(1,3,4,5) agonist) and SEW 2871 (an S1P(1) agonist) had no effects on pacemaker activity. Suramin (an S1P(3) antagonist) did not block the S1P-induced action on pacemaker currents. However, JTE-013 (an S1P(2) antagonist) blocked the S1P-induced action. RT-PCR revealed the presence of the S1P(2) in ICC. Calphostin C (a protein kinase C inhibitor), NS-398 (a cyclooxygenase-2 inhibitor), PD 98059 (a p42/44 inhibitor), or SB 203580 (a p38 inhibitor) had no effects on S1P-induced action. However, c-jun NH(2)-terminal kinase (JNK) inhibitor II suppressed S1P-induced action. External Ca(2+)-free solution or thapsigargin (a Ca(2+)-ATPase inhibitor of endoplasmic reticulum) suppressed action of S1P on ICC. In recording of intracellular Ca(2+) ([Ca(2+)](i)) concentration using fluo-4/AM S1P increased intensity of spontaneous [Ca(2+)](i) oscillations in ICC. These results suggest that S1P can modulate pacemaker activity of ICC through S1P(2) via regulation of external and internal Ca(2+) and mitogenactivated protein kinase activation.
Collapse
Affiliation(s)
- Young Dae Kim
- Department of Physiology, Department of Internal Medicine, College of Medicine, Chosun University, Gwangju 501-759,
Korea
| | - Kyoung Taek Han
- Department of Physiology, Department of Internal Medicine, College of Medicine, Chosun University, Gwangju 501-759,
Korea
| | - Jun Lee
- Department of Physiology, Department of Internal Medicine, College of Medicine, Chosun University, Gwangju 501-759,
Korea
| | - Chan Guk Park
- Department of Physiology, Department of Internal Medicine, College of Medicine, Chosun University, Gwangju 501-759,
Korea
| | - Man Yoo Kim
- Department of Physiology, Department of Internal Medicine, College of Medicine, Chosun University, Gwangju 501-759,
Korea
| | | | | | | | - Jae Yeoul Jun
- Department of Physiology, Department of Internal Medicine, College of Medicine, Chosun University, Gwangju 501-759,
Korea
| |
Collapse
|
13
|
Liu W, Lan T, Xie X, Huang K, Peng J, Huang J, Shen X, Liu P, Huang H. S1P2 receptor mediates sphingosine-1-phosphate-induced fibronectin expression via MAPK signaling pathway in mesangial cells under high glucose condition. Exp Cell Res 2012; 318:936-43. [PMID: 22406263 DOI: 10.1016/j.yexcr.2012.02.020] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2011] [Revised: 02/20/2012] [Accepted: 02/22/2012] [Indexed: 10/28/2022]
Abstract
Accumulation of extracellular matrix including fibronectin in mesangium is one of the major pathologic characteristics in diabetic nephropathy. In the current study, we explored role of sphingosine-1-phosphate (S1P) receptor in fibronectin expression and underlying molecular mechanism. Among five S1P receptors the mRNA level of S1P2 receptor was the most abundant in kidney of diabetic rats and mesangial cells under high glucose condition. S1P augmentation of fibronectin was significantly inhibited by S1P2 receptor antagonist JTE-013 and S1P2-siRNA. S1P-stimulated fibronectin expression was remarkably blocked by ERK1/2 inhibitor PD98059 and p38MAPK inhibitor SB203580. Phospho-ERK1/2 and phospho-p38MAPK level induced by S1P were markedly abrogated by JTE-013 and S1P2-siRNA. In conclusion, S1P2 receptor was significantly up-regulated under diabetic condition. S1P2 receptor mediated fibronectin expression through the activation of S1P-S1P2-MAPK (ERK1/2 and p38MAPK) axis in mesangial cells under high glucose condition, suggesting that it might be a potential therapeutic target for diabetic nephropathy treatment.
Collapse
Affiliation(s)
- Weihua Liu
- Laboratory of Pharmacology & Toxicology, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, China
| | | | | | | | | | | | | | | | | |
Collapse
|
14
|
Cross-talk between extracellular S1P/S1P2 and P42/44 MAPK in bcr/abl positive chronic myeloid leukemia cells. Chin J Cancer Res 2009. [DOI: 10.1007/s11670-009-0020-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022] Open
|
15
|
Affiliation(s)
- Yan Xu
- Department of Obstetrics and Gynecology, Indiana University, 975 W. Walnut St., IB355A, Indianapolis, IN 46202, USA.
| | | | | |
Collapse
|
16
|
Sphingosine-1-phosphate (S1P) is a novel fibrotic mediator in the eye. Exp Eye Res 2008; 87:367-75. [DOI: 10.1016/j.exer.2008.07.005] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2008] [Revised: 07/01/2008] [Accepted: 07/01/2008] [Indexed: 01/20/2023]
|
17
|
Peter BF, Lidington D, Harada A, Bolz HJ, Vogel L, Heximer S, Spiegel S, Pohl U, Bolz SS. Role of sphingosine-1-phosphate phosphohydrolase 1 in the regulation of resistance artery tone. Circ Res 2008; 103:315-24. [PMID: 18583713 PMCID: PMC2746908 DOI: 10.1161/circresaha.108.173575] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Sphingosine-1-phosphate (S1P), which mediates pleiotropic actions within the vascular system, is a prominent regulator of microvascular tone. By virtue of its S1P-degrading function, we hypothesized that S1P-phosphohydrolase 1 (SPP1) is an important regulator of tone in resistance arteries. Hamster gracilis muscle resistance arteries express mRNA encoding SPP1. Overexpression of SPP1 (via transfection of a SPP1(wt)) reduced resting tone, Ca2+ sensitivity, and myogenic vasoconstriction, whereas reduced SPP1 expression (antisense oligonucleotides) yielded the opposite effects. Expression of a phosphatase-dead mutant of SPP1 (SPP1(H208A)) had no effect on any parameter tested, suggesting that catalytic activity of SPP1 is critical. The enhanced myogenic tone that follows overexpression of S1P-generating enzyme sphingosine kinase 1 (Sk1(wt)) was functionally antagonized by coexpression with SPP1(wt) but not SPP1(H208A). SPP1 modulated vasoconstriction in response to 1 to 100 nmol/L exogenous S1P, a concentration range that was characterized as S1P2-dependent, based on the effect of S1P(2) inhibition by antisense oligonucleotides and 1 mumol/L JTE013. Inhibition of the cystic fibrosis transmembrane regulator (CFTR) (1) restored S1P responses that were attenuated by SPP1(wt) overexpression; (2) enhanced myogenic vasoconstriction; but (3) had no effect on noradrenaline responses. We conclude that SPP1 is an endogenous regulator of resistance artery tone that functionally antagonizes the vascular effects of both Sk1(wt) and S1P2 receptor activation. SPP1 accesses extracellular S1P pools in a manner dependent on a functional CFTR transport protein. Our study assigns important roles to both SPP1 and CFTR in the physiological regulation of vascular tone, which influences both tissue perfusion and systemic blood pressure.
Collapse
Affiliation(s)
| | - Darcy Lidington
- Institute of Physiology, Ludwig-Maximilians-University, Munich, Germany
- Department of Physiology and Heart & Stroke/Richard Lewar Centre of Excellence in Cardiovascular Research, University of Toronto, Canada
| | - Aki Harada
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University School of Medicine, Richmond
| | | | - Lukas Vogel
- Institute of Physiology, Ludwig-Maximilians-University, Munich, Germany
| | - Scott Heximer
- Department of Physiology and Heart & Stroke/Richard Lewar Centre of Excellence in Cardiovascular Research, University of Toronto, Canada
| | - Sarah Spiegel
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University School of Medicine, Richmond
| | - Ulrich Pohl
- Institute of Physiology, Ludwig-Maximilians-University, Munich, Germany
| | - Steffen-Sebastian Bolz
- Institute of Physiology, Ludwig-Maximilians-University, Munich, Germany
- Department of Physiology and Heart & Stroke/Richard Lewar Centre of Excellence in Cardiovascular Research, University of Toronto, Canada
| |
Collapse
|
18
|
Wang D, Zhao Z, Caperell-Grant A, Yang G, Mok SC, Liu J, Bigsby RM, Xu Y. S1P differentially regulates migration of human ovarian cancer and human ovarian surface epithelial cells. Mol Cancer Ther 2008; 7:1993-2002. [PMID: 18645009 PMCID: PMC2649755 DOI: 10.1158/1535-7163.mct-08-0088] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Epithelial ovarian cancer (EOC) arises from the epithelial layer covering the surface of ovaries and i.p. metastasis is commonly observed at diagnosis. Sphingosine-1-phosphate (S1P), a bioactive lipid signaling molecule, is potentially involved in EOC tumorigenesis. We have found that S1P is elevated in human EOC ascites. We show that physiologically relevant concentrations of S1P stimulate migration and invasion of EOC cells but inhibit migration of human ovarian surface epithelial (HOSE) cells. In addition, S1P inhibits lysophosphatidic acid (LPA)-induced cell migration in HOSE but not in EOC cells. We have provided the first line of evidence that the expression levels of S1P receptor subtypes are not the only determinants for how cells respond to S1P. Although S1P(1) is expressed and functional in HOSE cells, the inhibitory effect mediated by S1P(2) is dominant in those cells. The cellular preexisting stress fibers are also important determinants for the migratory response to S1P. Differential S1P-induced morphology changes are noted in EOC and HOSE cells. Preexisting stress fibers in HOSE cells are further enhanced by S1P treatment, resulting in the negative migratory response to S1P. By contrast, EOC cells lost stress fibers and S1P treatment induces filopodium-like structures at cell edges, which correlates with increased cell motility. In addition, inhibition of the protein kinase C pathway is likely to be involved in the inhibitory effect of S1P on LPA-induced cell migration in HOSE cells. These findings are important for the development of new therapeutics targeting S1P and LPA in EOC.
Collapse
Affiliation(s)
- Dongmei Wang
- Department of Obstetrics and Gynecology, Indiana University, 975 West Walnut Street IB355A, Indianapolis, IN 46202, USA
| | | | | | | | | | | | | | | |
Collapse
|
19
|
Li QF, Wu CT, Guo Q, Wang H, Wang LS. Sphingosine 1-phosphate induces Mcl-1 upregulation and protects multiple myeloma cells against apoptosis. Biochem Biophys Res Commun 2008; 371:159-62. [DOI: 10.1016/j.bbrc.2008.04.037] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2008] [Accepted: 04/04/2008] [Indexed: 01/11/2023]
|
20
|
Abstract
Most of the previous work on the sphingolipid ceramide has been devoted to its function as an apoptosis inducer. Recent studies, however, have shown that in stem cells, ceramide has additional nonapoptotic functions. In this article, ceramide signaling will be reviewed in light of 'systems interface biology': as an interconnection of sphingolipid metabolism, membrane biophysics and cell signaling. The focus will be on the metabolic interconversion of ceramide and sphingomyelin or sphingosine-1-phosphate. Lipid rafts and sphingolipid-induced protein scaffolds will be discussed as a membrane interface for lipid-controlled cell signaling. Ceramide/sphingomyelin and ceramide/sphingosine-1-phosphate-interdependent cell-signaling pathways are significant for the regulation of cell polarity, apoptosis and/or proliferation, and as novel pharmacologic targets in cancer and stem cells.
Collapse
Affiliation(s)
- Erhard Bieberich
- Institute of Molecular Medicine & Genetics, School of Medicine, Medical College of Georgia, 1120 15th Street, Room CB-2803, Augusta, GA 30912, USA
| |
Collapse
|
21
|
Bieberich E. Smart drugs for smarter stem cells: making SENSe (sphingolipid-enhanced neural stem cells) of ceramide. Neurosignals 2008; 16:124-39. [PMID: 18253053 DOI: 10.1159/000111558] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Ceramide and its derivative sphingosine-1-phosphate (S1P) are important signaling sphingolipids for neural stem cell apoptosis and differentiation. Most recently, our group has shown that novel ceramide analogs can be used to eliminate teratoma (stem cell tumor)-forming cells from a neural stem cell graft. In new studies, we found that S1P promotes survival of specific neural precursor cells that undergo differentiation to cells expressing oligodendroglial markers. Our studies suggest that a combination of novel ceramide and S1P analogs eliminates tumor-forming stem cells and at the same time, triggers oligodendroglial differentiation. This review discusses recent studies on the function of ceramide and S1P for the regulation of apoptosis, differentiation, and polarity in stem cells. We will also discuss results from ongoing studies in our laboratory on the use of sphingolipids in stem cell therapy.
Collapse
Affiliation(s)
- Erhard Bieberich
- Program in Developmental Neurobiology, Institute of Molecular Medicine and Genetics, School of Medicine, Medical College of Georgia, Augusta, GA 30912, USA.
| |
Collapse
|
22
|
Roztocil E, Nicholl SM, Davies MG. Sphingosine-1-phosphate-induced oxygen free radical generation in smooth muscle cell migration requires Galpha12/13 protein-mediated phospholipase C activation. J Vasc Surg 2007; 46:1253-1259. [PMID: 18155002 PMCID: PMC2607047 DOI: 10.1016/j.jvs.2007.08.013] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2007] [Revised: 07/28/2007] [Accepted: 08/05/2007] [Indexed: 10/22/2022]
Abstract
BACKGROUND Sphingosine-1-phosphate (S-1-P) is a bioactive sphingolipid that stimulates the migration of vascular smooth muscle cell (VSMC) through G-protein coupled receptors; it has been shown to activate reduced nicotinamide dinucleotide phosphate hydrogen (NAD[P]H) oxidase. The role of phospholipase C (PLC) in oxygen free radical generation, and the regulation of VSMC migration in response to S-1-P, are poorly understood. METHODS Rat arterial VSMC were cultured in vitro. Oxygen free radical generation was measured by fluorescent redox indicator assays in response to S-1-P (0.1microM) in the presence and absence of the active PLC inhibitor (U73122; U7, 10nM) or its inactive analog U73343 (InactiveU7, 10nM). Activation of PLC was assessed by immunoprecipitation and Western blotting for the phosphorylated isozymes (beta and gamma). Small interfering (si) RNA to the G-proteins Galphai, Galphaq, and Galpha12/13 was used to downregulate specific proteins. Statistics were by one-way analysis of variance (n = 6). RESULTS S-1-P induced time-dependent activation of PLC-beta and PLC-gamma; PLC-beta but not PLC-gamma activation was blocked by U7 but not by InactiveU7. PLC-beta activation was Galphai-independent (not blocked by pertussis toxin, a Galphai inhibitor, or Galphai2 and Galphai3 siRNA) and Galphaq-independent (not blocked by glycoprotein [GP] 2A, a Galphaq inhibitor, or Galphaq siRNA). PLC-beta activation and cell migration was blocked by siRNA to Galpha12/13. Oxygen free radical generation induced by S-1-P, as measured by dihydroethidium staining, was significantly inhibited by U7 but not by InactiveU7. Inhibition of oxygen free radicals with the inhibitor diphenyleneiodonium resulted in decreased cell migration to S-1-P. VSMC mitogen-activated protein kinase activation and VSMC migration in response to S-1-P was inhibited by PLC- inhibition. CONCLUSION S-1-P induces oxygen free radical generation through a Galpha12/13, PLC-beta-mediated mechanism that facilitates VSMC migration. To our knowledge, this is the first description of PLC-mediated oxygen free radical generation as a mediator of S-1-P VSMC migration and illustrates the need for the definition of cell signaling to allow targeted strategies in molecular therapeutics for restenosis.
Collapse
MESH Headings
- Animals
- Cell Movement/drug effects
- Cells, Cultured
- Enzyme Activation
- Estrenes/pharmacology
- GTP-Binding Protein alpha Subunits, G12-G13/antagonists & inhibitors
- GTP-Binding Protein alpha Subunits, G12-G13/genetics
- GTP-Binding Protein alpha Subunits, G12-G13/metabolism
- Lysophospholipids/metabolism
- MAP Kinase Signaling System
- Mitogen-Activated Protein Kinase 1/metabolism
- Mitogen-Activated Protein Kinase 3/metabolism
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/enzymology
- Muscle, Smooth, Vascular/metabolism
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/enzymology
- Myocytes, Smooth Muscle/metabolism
- NADPH Oxidases/antagonists & inhibitors
- NADPH Oxidases/metabolism
- Onium Compounds/pharmacology
- Pertussis Toxin/pharmacology
- Phosphodiesterase Inhibitors/pharmacology
- Phospholipase C beta/antagonists & inhibitors
- Phospholipase C beta/metabolism
- Phospholipase C gamma/metabolism
- Phosphorylation
- Pyrrolidinones/pharmacology
- RNA Interference
- RNA, Small Interfering/metabolism
- Rats
- Rats, Sprague-Dawley
- Reactive Oxygen Species/metabolism
- Signal Transduction/drug effects
- Sphingosine/analogs & derivatives
- Sphingosine/metabolism
- Time Factors
- p38 Mitogen-Activated Protein Kinases/metabolism
Collapse
Affiliation(s)
- Eliza Roztocil
- Vascular Biology and Therapeutics Program, Department of Surgery, University of Rochester, Rochester, NY 14642, USA
| | | | | |
Collapse
|
23
|
Watterson KR, Lanning DA, Diegelmann RF, Spiegel S. Regulation of fibroblast functions by lysophospholipid mediators: Potential roles in wound healing. Wound Repair Regen 2007; 15:607-16. [DOI: 10.1111/j.1524-475x.2007.00292.x] [Citation(s) in RCA: 92] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
|
24
|
Kawanabe T, Kawakami T, Yatomi Y, Shimada S, Soma Y. Sphingosine 1-phosphate accelerates wound healing in diabetic mice. J Dermatol Sci 2007; 48:53-60. [PMID: 17643267 DOI: 10.1016/j.jdermsci.2007.06.002] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2007] [Revised: 06/12/2007] [Accepted: 06/12/2007] [Indexed: 11/28/2022]
Abstract
BACKGROUND Blood platelets store sphingosine 1-phosphate (S1P) abundantly and release this bioactive lipid extracellularly. S1P acts as an intercellular mediator through interaction with the endothelial differentiation gene (EDG)/S1P family of G protein-coupled receptors. Of the EDG family S1P receptors, EDG-5 (S1P2) is inhibited in migration induced by S1P. Diabetes impairs numerous aspects of tissue repair. Failure of wound angiogenesis is known to delay diabetic wound healing. OBJECTIVES We examined whether S1P subcutaneous injection could improve the healing of full-thickness skin wounds in healthy and diabetic mice. We further determine if the combined S1P and EDG-5 (S1P2) antagonist injection in diabetic mice could affect wound healing. Finally, we examined the histopathological findings of the wound following S1P injection in diabetic mice. METHODS Eight- to 10-week-old BALA/c mice, diabetic db/db mice and Wister rats were used for the studies. A full-thickness wound was made on the dorsal skin of the healthy and diabetic mice. Either 10 microM or 100 microM of S1P or vehicle control (BSA/PBS) was injected into the wound bed every day. We calculated the wound area after each injection. EDG-5 (S1P2) antagonist (JTE-013) or vehicle (DMSO) was then injected in addition to the S1P around the dorsal wound of diabetic mice and the wound diameter was measured. Wound tissue samples were excised following injection for histopathological examination. RESULTS Wound area in normal BALA/c mice did not significantly decrease upon S1P injection compared to S1P-untreated controls. S1P injection alone showed significant promotion of wound healing in diabetic mice compared to no S1P treatment. The combination of S1P and EDG-5 (S1P2) receptor antagonist administration induced maximal wound healing in diabetic mice. Histopathological examination revealed that S1P induces neo-vascularization potential in rats and diabetic mice wound. CONCLUSIONS S1P injection in diabetic mice significantly accelerated cutaneous wound healing in the neo-vascularization process. The results demonstrate that S1P affects and sustains all key cellular processes responsible for wound repair and point to a unique potential for this molecule in the therapy of diabetic wounds, particularly as an angiogenic agent in treatment of diabetic wounds.
Collapse
MESH Headings
- Animals
- Diabetes Mellitus, Experimental/pathology
- Diabetes Mellitus, Experimental/physiopathology
- Disease Models, Animal
- Drug Therapy, Combination
- Injections, Subcutaneous
- Lysophospholipids/administration & dosage
- Lysophospholipids/pharmacology
- Lysophospholipids/physiology
- Male
- Mice
- Mice, Inbred BALB C
- Mice, Inbred NOD
- Neovascularization, Physiologic/physiology
- Pyrazoles/administration & dosage
- Pyrazoles/pharmacology
- Pyridines/administration & dosage
- Pyridines/pharmacology
- Rats
- Rats, Wistar
- Receptors, Lysosphingolipid/antagonists & inhibitors
- Skin/blood supply
- Skin/pathology
- Skin/physiopathology
- Sphingosine/administration & dosage
- Sphingosine/analogs & derivatives
- Sphingosine/pharmacology
- Sphingosine/physiology
- Wound Healing/drug effects
- Wound Healing/physiology
Collapse
Affiliation(s)
- Takeshi Kawanabe
- Department of Dermatology, St. Marianna University School of Medicine, Japan
| | | | | | | | | |
Collapse
|
25
|
Aoki S, Yatomi Y, Shimosawa T, Yamashita H, Kitayama J, Tsuno NH, Takahashi K, Ozaki Y. The suppressive effect of sphingosine 1-phosphate on monocyte-endothelium adhesion may be mediated by the rearrangement of the endothelial integrins alpha(5)beta(1) and alpha(v)beta(3). J Thromb Haemost 2007; 5:1292-301. [PMID: 17403093 DOI: 10.1111/j.1538-7836.2007.02559.x] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
BACKGROUND Sphingosine 1-phosphate (S1P), known to play important roles in vascular biology, is a bioactive lysophospholipid mediator that maintains endothelial integrity via its cell-surface receptors (S1Ps). In this in vitro study, we aimed to examine the role of S1P in monocyte-endothelium adhesion, which is an important event in the pathophysiology of atherosclerosis. METHODS AND RESULTS S1P pretreatment of human umbilical vein endothelial cells (ECs), but not U937 cells, effectively suppressed U937-EC adhesion independently from the expression of adhesion molecules, namely ICAM-1, VCAM-1, and E-selectin. This S1P-induced suppressive effect was inhibited by the blockage of S1P(1) and S1P(3) receptors and the specific inhibitors of G(i) protein, Src family proteins, phosphatidylinositol 3-kinase, and Rac1, indicating involvement of these key downstream pathways. Moreover, the RGD peptide and antibodies, which neutralize adhesion via alpha(5)beta(1) and alpha(v)beta(3), effectively inhibited U937-EC adhesion with a degree similar to S1P pretreatment. Both an adhesion assay and flow-cytometric analysis demonstrated that U937 cells adhered through integrins alpha(5)beta(1) and alpha(v)beta(3) expressed on the apical surface of monolayer ECs, and S1P shifted the localization of these integrins from the apical surface to the basal surface. CONCLUSIONS From the present results, we propose that S1P may contribute to the maintenance of vascular integrity and the regulation of atherogenesis through the rearrangement of endothelial integrins.
Collapse
Affiliation(s)
- S Aoki
- Department of Clinical Laboratory Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | | | | | | | | | | | | | | |
Collapse
|
26
|
Nodai A, Machida T, Izumi S, Hamaya Y, Kohno T, Igarashi Y, Iizuka K, Minami M, Hirafuji M. Sphingosine 1-phosphate induces cyclooxygenase-2 via Ca2+-dependent, but MAPK-independent mechanism in rat vascular smooth muscle cells. Life Sci 2007; 80:1768-76. [PMID: 17382352 DOI: 10.1016/j.lfs.2007.02.008] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2006] [Revised: 01/12/2007] [Accepted: 02/07/2007] [Indexed: 02/07/2023]
Abstract
The effects of sphingosine 1-phosphate (S1P) on prostaglandin I(2) (PGI(2)) production and cyclooxygenase (COX) expression in cultured rat vascular smooth muscle cells (VSMCs) were investigated. S1P stimulated PGI(2) production in a concentration-dependent manner, which was completely suppressed by NS-398, a selective COX-2 inhibitor, as determined by radioimmunoassay. S1P stimulated COX-2 protein and mRNA expressions in a concentration- and time-dependent manner, while it had no effect on COX-1 expression. S1P(2) and S1P(3) receptors mRNA were abundantly expressed in rat VSMCs. Suramin, an antagonist of S1P(3) receptor, almost completely inhibited S1P-induced COX-2 expression. Pretreatment of VSMCs with pertussis toxin (PTX) partially, but significantly inhibited S1P-induced PGI(2) production and COX-2 expression. S1P also activated extracellular signal-regulated kinase (ERK) and p38 mitogen-activated protein kinase (MAPK). However, neither PD 98059, a selective inhibitor of ERK activation, nor SB 203580, a selective inhibitor of p38 MAPK, had a significant inhibitory effect on S1P-induced COX-2 expression, suggesting that the MAPK activation does not play main roles in S1P-induced COX-2 induction. S1P-induced COX-2 expression was inhibited by PP2, an inhibitor of Src-family tyrosine kinase, Ca(2+) depletion, and GF 109203X, an inhibitor of protein kinase C (PKC). These results suggest that S1P stimulates COX-2 induction in rat VSMCs through mechanisms involving Ca(2+)-dependent PKC and Src-family tyrosine kinase activation via S1P(3) receptor coupled to PTX-sensitive and -insensitive G proteins.
Collapse
MESH Headings
- Animals
- Calcium/metabolism
- Cells, Cultured
- Cyclooxygenase 2/drug effects
- Cyclooxygenase 2/genetics
- Cyclooxygenase 2/metabolism
- Cyclooxygenase Inhibitors/pharmacology
- Epoprostenol/biosynthesis
- Epoprostenol/metabolism
- Lysophospholipids/pharmacology
- Mitogen-Activated Protein Kinases/antagonists & inhibitors
- Mitogen-Activated Protein Kinases/metabolism
- Muscle, Smooth, Vascular/cytology
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/enzymology
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/enzymology
- Nitrobenzenes/pharmacology
- Pertussis Toxin/pharmacology
- Protein Kinase C/antagonists & inhibitors
- Protein Kinase C/metabolism
- Protein Kinase Inhibitors/pharmacology
- RNA, Messenger/metabolism
- Rats
- Rats, Wistar
- Receptors, Lysosphingolipid/agonists
- Receptors, Lysosphingolipid/genetics
- Receptors, Lysosphingolipid/metabolism
- Sphingosine/analogs & derivatives
- Sphingosine/pharmacology
- Sulfonamides/pharmacology
- src-Family Kinases/antagonists & inhibitors
- src-Family Kinases/metabolism
Collapse
Affiliation(s)
- Akiko Nodai
- Department of Pharmacology, Faculty of Pharmaceutical Sciences, Health Sciences University of Hokkaido, Ishikari-Tobetsu, Hokkaido 061-0293, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Nakamura T, Yonesu K, Mizuno Y, Suzuki C, Sakata Y, Takuwa Y, Nara F, Satoh S. Synthesis and SAR studies of a novel class of S1P1 receptor antagonists. Bioorg Med Chem 2007; 15:3548-64. [PMID: 17379528 DOI: 10.1016/j.bmc.2007.02.048] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2006] [Revised: 02/22/2007] [Accepted: 02/23/2007] [Indexed: 10/23/2022]
Abstract
A series of Sodium 4-[(4-butoxyphenyl)thio]-2'-substituted-1,1'-biphenyl-3- sulfonates were identified as functional sphingosine-1-phosphate (S1P) antagonists with selectivity for the S1P(1) receptor subtype starting from chemical lead 2, which was found while screening our in-house compound library. We performed chemical modifications on each regional structure of compound 2, for example, on the three ring compartments, the benzyl substituents, and the long alkyl chain part. The introduction of a biphenyl skeletal structure and the installation of a hydroxyl group onto the terminal carbon in the side-chain region resulted in the potent derivative 35c, which showed >500-fold more potent S1P(1) inhibitory activity than lead compound 2. We report herein the synthesis and structure-activity relationships of structurally novel S1P(1) receptor antagonists.
Collapse
Affiliation(s)
- Tsuyoshi Nakamura
- Medicinal Chemistry Research Laboratories, Sankyo Co., Ltd, 1-2-58 Hiromachi, Tokyo 140-8710, Japan
| | | | | | | | | | | | | | | |
Collapse
|
28
|
Yamashita H, Kitayama J, Shida D, Yamaguchi H, Mori K, Osada M, Aoki S, Yatomi Y, Takuwa Y, Nagawa H. Sphingosine 1-phosphate receptor expression profile in human gastric cancer cells: differential regulation on the migration and proliferation. J Surg Res 2005; 130:80-7. [PMID: 16183075 DOI: 10.1016/j.jss.2005.08.004] [Citation(s) in RCA: 65] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2005] [Revised: 07/26/2005] [Accepted: 08/01/2005] [Indexed: 01/12/2023]
Abstract
INTRODUCTION Sphingosine 1-phosphate (S1P) is a bioactive lysophospholipid, derived from activated platelet, that is known to induce diverse cellular responses through at least five G-protein-coupled receptors on various cell types. Abnormal platelet and coagulation activation is often seen in patients with gastric cancer. However, neither the effects of this platelet-derived mediator S1P nor the distribution of S1P receptors on the gastric cancer cell are fully understood. The aim of this study was to examine the possible role of S1P and its receptors in the progression of gastric cancer. MATERIALS AND METHODS We characterized the expression profiles of S1P receptors in nine human gastric cancer cell lines and evaluated the relationship between the responses to S1P and its receptor expression on cell migration by modified Boyden chamber and cell proliferation by MTS assay. RESULTS Northern blotting analysis has revealed that S1P2 was expressed in all gastric cancer cell lines to varying degrees, and S1P3 was expressed in four cell lines. S1P1 expression was weak, and no significant expression of either S1P4 or S1P5 was detected. The addition of S1P markedly stimulated the migration of MKN1 and HCG-27 that dominantly expressed S1P3, and the effect was potently inhibited by pertussis toxin or wortmannin. In contrast, SIP significantly inhibited the migration of AZ-521 that expressed S1P2 exclusively. This indicates that the balance between S1P2- and S1P3-mediated signals might be critical in determining the metastatic response of gastric cancer cells to S1P. S1P elicited weak but significant antiproliferative effects on all of the three cell lines, although the effects were not major. In these cells, S1P induced extracellular signal-regulated kinase (ERK) phosphorylation with transient Akt dephosphorylation that may cause the weak effects on proliferation. CONCLUSIONS Our results suggest that the S1P receptor expression may critically determine the biological behavior of gastric cancers and thus therapeutic interventions directed at each S1P receptor might be clinically effective in preventing metastasis in gastric cancer.
Collapse
Affiliation(s)
- Hiroharu Yamashita
- Department of Surgical Oncology, University of Tokyo Graduate School of Medicine, Tokyo, Japan.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Watterson KR, Ratz PH, Spiegel S. The role of sphingosine-1-phosphate in smooth muscle contraction. Cell Signal 2005; 17:289-98. [PMID: 15567060 DOI: 10.1016/j.cellsig.2004.09.013] [Citation(s) in RCA: 94] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2004] [Revised: 09/02/2004] [Accepted: 09/02/2004] [Indexed: 01/12/2023]
Abstract
Sphingosine-1-phosphate (S1P) is a bioactive sphingolipid metabolite that is known to mediate diverse cellular responses including cell growth, survival, and migration. Most of these effects have been attributed to its binding to a specific subfamily of G protein-coupled receptors (GPCR), namely S1P(1-5). Recent studies have suggested that S1P also plays a prominent role in the contraction of various types of smooth muscle. This review provides a brief overview of its role in this process and also highlights how S1P-dependent signaling serves as an important regulator of smooth muscle contraction.
Collapse
Affiliation(s)
- Kenneth R Watterson
- Department of Biochemistry, Virginia Commonwealth University School of Medicine, 1101 E. Marshall Street, P.O. Box 980614, Sanger Hall, Room 2-011, Richmond, VA 23298-0614, USA
| | | | | |
Collapse
|
30
|
Murakami T, Furusawa K, Tamai T, Yoshikai K, Nishikawa M. Synthesis and biological properties of novel sphingosine derivatives. Bioorg Med Chem Lett 2005; 15:1115-9. [PMID: 15686924 DOI: 10.1016/j.bmcl.2004.12.010] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2004] [Revised: 12/02/2004] [Accepted: 12/06/2004] [Indexed: 11/20/2022]
Abstract
Sphingosine-1-phosphate (S-1P) derivatives such as threo-(2S,3S)-analogues, which are C-3 stereoisomers of natural erythro-(2S,3R)-S-1P, have been synthesized starting from l-serine or (1S,2S)-2-amino-1-aryl-1,3-propanediols (6). threo-(1S,2R)-2-Amino-1-aryl-3-bromopropanols (HBr salt) have also been prepared from 6. The threo-S-1Ps and the threo-amino-bromide derivatives have shown potent inhibitory activity against Ca(2+) ion mobilization in HL60 cells induced by erythro-S-1P, suggesting that these compounds would compete with cell surface EDG/S1P receptors.
Collapse
Affiliation(s)
- Teiichi Murakami
- National Institute of Advanced Industrial Science and Technology (AIST), Tsukuba Central 5, Tsukuba, Ibaraki 305-8565, Japan.
| | | | | | | | | |
Collapse
|
31
|
Houjou T, Yamatani K, Imagawa M, Shimizu T, Taguchi R. A shotgun tandem mass spectrometric analysis of phospholipids with normal-phase and/or reverse-phase liquid chromatography/electrospray ionization mass spectrometry. RAPID COMMUNICATIONS IN MASS SPECTROMETRY : RCM 2005; 19:654-666. [PMID: 15700236 DOI: 10.1002/rcm.1836] [Citation(s) in RCA: 138] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
Electrospray ionization mass spectrometry is used in lipidomics studies. The present research established a top-down liquid chromatography/electrospray ionization tandem mass spectrometry (LC/ESI-MS/MS) shotgun analysis method for phospholipids (PLs) using a normal-phase column or a C30 reverse-phase column with the data-dependent MS/MS scanning mode. A normal-phase column can separate most of the major different classes of PLs. By using LC/ESI-MS/MS with a normal-phase column, approximately 50 molecular species were identified in a PL mixture from rat liver. When the reverse-phase column was used, the PLs could be separated depending on their hydrophobicity, essentially the length of their fatty acyl chains and the number of unsaturated bonds in them. The LC/ESI-MS/MS method using a C30 reverse-phase column was applied to phosphatidylcholine (PC) and phosphatidylethanolamine (PE) mixtures as test samples. Molecular species with the same molecular mass but with different pairs of fatty acyl chains were separately identified. As a result, about 60 PC and 50 PE species were identified. PLs from rat liver were subjected to LC/ESI-MS/MS using the C30 reverse-phase column and about 110 molecular species were identified. Off-line two-dimensional LC/ESI-MS/MS with the normal-phase and C30 reverse-phase columns allowed more accurate identification of molecular species by using one-dimensional C30 reverse-phase LC/ESI-MS/MS analysis of the collected fractions.
Collapse
Affiliation(s)
- Toshiaki Houjou
- Graduate School of Pharmaceutical Sciences, Nagoya City University, Mizuho-ku, Nagoya City, Aichi 467-8603, Japan
| | | | | | | | | |
Collapse
|
32
|
Alewijnse AE, Peters SLM, Michel MC. Cardiovascular effects of sphingosine-1-phosphate and other sphingomyelin metabolites. Br J Pharmacol 2004; 143:666-84. [PMID: 15504747 PMCID: PMC1575924 DOI: 10.1038/sj.bjp.0705934] [Citation(s) in RCA: 112] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Upon various stimuli, cells metabolize sphingomyelin from the cellular plasma membrane to form sphingosylphosphorylcholine (SPC) or ceramide. The latter can be further metabolized to sphingosine and then sphingosine-1-phosphate (S1P). Apart from local formation, S1P and SPC are major constituents of blood plasma. All four sphingomyelin metabolites (SMM) can act upon intracellular targets, and at least S1P and probably also SPC can additionally act upon G-protein-coupled receptors. While the molecular identity of the SPC receptors remains unclear, several subtypes of S1P receptors have been cloned and their distribution in cardiovascular tissues is described. In the heart SMM can alter intracellular Ca(2+) release, particularly via the ryanodine receptor, and conductance of various ion channels in the plasma membrane, particularly I(K(Ach)). While the various SMM differ somewhat in their effects, the above alterations of ion homeostasis result in reduced cardiac function in most cases, and ceramide and/or sphingosine may be the mediators of the negative inotropic effects of tumour necrosis factor. In the vasculature, SMM mainly act as acute vasoconstrictors in most vessels, but ceramide can be a vasodilator. SMM-induced vasoconstriction involves mobilization of Ca(2+) from intracellular stores, influx of extracellular Ca(2+) via L-type channels and activation of a rho-kinase. Extended exposure to SMM, particularly S1P, promotes several stages of the angiogenic process like endothelial cell activation, migration, proliferation, tube formation and vascular maturation. We propose that SMM are an important class of endogenous modulators of cardiovascular function.
Collapse
Affiliation(s)
- Astrid E Alewijnse
- Department of Pharmacology and Pharmacotherapy, University of Amsterdam, The Netherlands
| | - Stephan L M Peters
- Department of Pharmacology and Pharmacotherapy, University of Amsterdam, The Netherlands
| | - Martin C Michel
- Department of Pharmacology and Pharmacotherapy, University of Amsterdam, The Netherlands
- Author for correspondence:
| |
Collapse
|
33
|
Yamada M, Banno Y, Takuwa Y, Koda M, Hara A, Nozawa Y. Overexpression of phospholipase D prevents actinomycin D-induced apoptosis through potentiation of phosphoinositide 3-kinase signalling pathways in Chinese-hamster ovary cells. Biochem J 2004; 378:649-56. [PMID: 14640974 PMCID: PMC1223985 DOI: 10.1042/bj20031398] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2003] [Revised: 11/17/2003] [Accepted: 11/25/2003] [Indexed: 12/16/2022]
Abstract
To examine the roles of PLD (phospholipase D) in the regulation of the apoptotic process, PLD1 and PLD2 were stably overexpressed in S1P3-CHO cells [CHO (Chinese-hamster ovary) cells expressing the S1P (sphingosine 1-phosphate) receptor S1P3]. Treatment of S1P3-CHO cells with ActD (actinomycin D) induced apoptosis, as shown by the occurrence of nuclear fragmentation and the caspase-dependent proteolytic cleavage of PARP [poly(ADP-ribose) polymerase] and protein kinase Cd. Overexpression of either PLD1 or PLD2 protected S1P3-CHO cells from ActD-induced apoptosis, as demonstrated by an increased number of viable cells and inhibition of PARP and protein kinase Cd cleavage. However, in the early phase of apoptosis, ActD induced an increase in PLD activity and activation of key factors in the cell-survival signalling pathways, such as PI3K (phosphoinositide 3-kinase), Akt, p70S6K (p70 S6 kinase) and ERK (extracellular-signal-regulated kinase). Furthermore, the ActD-induced activation of these survival signalling enzymes was potentiated by overexpression of either PLD1 or PLD2. The PI3K inhibitor LY294002 inhibited the ActD-induced activation of Akt and p70S6K, and completely abolished the effects of PLD1 or PLD2, whereas inhibition of ERK activity by the MEK inhibitor U0126 had a milder effect. The ActD-induced activation of p70S6K and ERKs was blocked by 1-butanol, but not by t-butanol; similar to S1P, exogenous PLD suppressed the ActD-induced events in the apoptosis signalling pathways. These results show that, in S1P3-CHO cells, increased expression of PLDs prevents ActD-induced apoptosis by enhanced activation of the PI3K signalling pathways.
Collapse
Affiliation(s)
- Momoko Yamada
- Department of Biochemistry, Gifu Pharmaceutical University, Mitahora, Gifu, Japan
| | | | | | | | | | | |
Collapse
|
34
|
Hanada M, Feng J, Hemmings BA. Structure, regulation and function of PKB/AKT--a major therapeutic target. BIOCHIMICA ET BIOPHYSICA ACTA-PROTEINS AND PROTEOMICS 2004; 1697:3-16. [PMID: 15023346 DOI: 10.1016/j.bbapap.2003.11.009] [Citation(s) in RCA: 547] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/12/2003] [Accepted: 11/12/2003] [Indexed: 12/20/2022]
Abstract
Protein phosphorylation and dephosphorylation play a major role in intracellular signal transduction activated by extracellular stimuli. Protein kinase B (PKB/Akt) is a central player in the signal transduction pathways activated in response to growth factors or insulin and is thought to contribute to several cellular functions including nutrient metabolism, cell growth and apoptosis. Recently, several significant publications have described novel mechanisms used to regulate PKB. Since the alteration of PKB activity is associated with several human diseases, including cancer and diabetes, understanding PKB regulation is an important task if we are to develop successful therapeutics.
Collapse
Affiliation(s)
- Masahito Hanada
- Friedrich Miescher Institute for Biomedical Research, Maulbeerstrasse 66, Basel CH-4058, Switzerland
| | | | | |
Collapse
|
35
|
Dawson J, Hotchin N, Lax S, Rumsby M. Lysophosphatidic acid induces process retraction in CG-4 line oligodendrocytes and oligodendrocyte precursor cells but not in differentiated oligodendrocytes. J Neurochem 2004; 87:947-57. [PMID: 14622125 DOI: 10.1046/j.1471-4159.2003.02056.x] [Citation(s) in RCA: 39] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Lysophosphatidic acid is a growth factor-like signalling phospholipid. We demonstrate here that lysophosphatidic acid induces process retraction in central glia-4 cells and oligodendrocyte precursors. This lysophosphatidic acid effect is rapid and concentration-dependent and results in cell rounding. It is inhibited by pre-treatment of cells with C3 exoenzyme, a specific inhibitor of Rho, or with Y-27632, a specific inhibitor of ROCK, a downstream kinase of Rho. Processes of differentiated central glia-4 oligodendrocytes were insensitive to lysophosphatidic acid treatment but cell bodies became phase dark, indicating cell spreading on the poly-l-lysine substratum. RT-PCR and Western blot analyses indicate that oligodendrocyte precursors and mature oligodendrocytes express mRNA and protein for LPA1, one of several LPA receptors. Thus lysophosphatidic acid may be signalling to Rho and stimulating actomyosin contraction in precursor oligodendrocytes by this family of receptors. The results show that lysophosphatidic acid signalling pathways influence retraction of processes in oligodendrocyte precursors but that this effect changes as oligodendrocytes differentiate.
Collapse
Affiliation(s)
- John Dawson
- Department of Biology, University of York, York, UK.
| | | | | | | |
Collapse
|
36
|
Takeya H, Gabazza EC, Aoki S, Ueno H, Suzuki K. Synergistic effect of sphingosine 1-phosphate on thrombin-induced tissue factor expression in endothelial cells. Blood 2003; 102:1693-700. [PMID: 12730100 DOI: 10.1182/blood-2002-11-3607] [Citation(s) in RCA: 53] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Sphingosine 1-phosphate (S1P), a bioactive lipid, is produced and stored in platelets and is released from activated platelets during blood coagulation activation. Thrombin, which is also generated during blood coagulation, has been shown to induce tissue factor (TF), the initiator of blood coagulation, in endothelial cells (ECs); however, the effect of S1P on this process is not evaluated. Here we demonstrated that S1P strongly potentiated thrombin-induced TF expression in ECs and that S1P itself did not induce TF expression. Among signaling lipids, platelet-activating factor slightly enhanced thrombin-induced TF expression; other lipids, including lysophosphatidic acid, lysophosphatidylcholine, sphingosine, and C2-ceramide exert no effect on TF expression. S1P enhanced TF expression at the transcriptional level, possibly via promoting the activation of transcription factors nuclear factor-kappaB (NF-kappaB) and Egr-1. Thrombin weakly and S1P strongly activated extracellular signal-regulated kinase 1/2 (ERK1/2) mitogen-activated protein (MAP) kinase and, in the presence of both stimulants, enhanced and sustained activation of this kinase was observed. The ERK1/2-specific inhibitor PD98059 significantly inhibited enhanced TF expression induced by both stimulants but only weakly inhibited thrombin-induced TF expression, thus indicating the requirement of the ERK1/2 pathway in synergistic induction of TF expression. In addition, we found that thrombin and S1P rapidly up-regulated the expression of S1P receptors, endothelial differentiation gene-1 (EDG-1) and EDG-3, thereby suggesting that the effect of S1P on TF expression and other EC functions may be enhanced by thrombin and S1P itself. The present data reveal the synergistic effect of S1P on thrombin-induced TF expression in ECs, which may promote further thrombin and S1P generation, thus propagating a positive feedback reaction.
Collapse
Affiliation(s)
- Hiroyuki Takeya
- Department of Biochemistry, University of Occupational and Environmental Health, School of Medicine, Kitakyushu 807-8555, Japan.
| | | | | | | | | |
Collapse
|
37
|
Villullas IR, Smith AJ, Heavens RP, Simpson PB. Characterisation of a sphingosine 1-phosphate-activated Ca2+ signalling pathway in human neuroblastoma cells. J Neurosci Res 2003; 73:215-26. [PMID: 12836164 DOI: 10.1002/jnr.10636] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Sphingosine 1-phosphate (S1P) has assumed great importance within neuroscience research because of putative links between S1P-sensitive Edg receptors and neuroregeneration, cell survival, and alterations in neurite outgrowth. In the present study, we examined the mechanisms by which the endogenous complement of S1P-sensitive human Edg receptors can elevate Ca(2+) in the human neuroblastoma cell line, SH-SY5Y. Reverse transcriptase-polymersase chain reaction (RT-PCR) confirmed the expression of mRNA for Edg 3, 5, and 8 subtypes of S1P-responsive Edg receptors in SH-SY5Y cells. Neither S1P nor the muscarinic agonist methacholine were able to cause a change in SH-SY5Y cell morphology, whereas retinoic acid caused a range of changes, including an increase in neurite outgrowth, under similar test conditions. Stimulation with S1P resulted in a slowly rising increase in cytosolic Ca(2+) levels. These responses were dependent upon inositol-1,4,5-trisphosphate receptors, thapsigargin-sensitive endoplasmic reticulum, and also intact functional mitochondria. S1P-evoked Ca(2+) responses were similar in mechanism to those of methacholine, which activated a much faster responding, larger amplitude Ca(2+) response. These studies indicate that in an endogenous human expression system, S1P appears to be an efficacious agonist of Edg receptors. Despite its slow time course of response, S1P appears to activate the same single Ca(2+) store in SH-SY5Y cells as is activated by methacholine and other G protein coupled receptors.
Collapse
Affiliation(s)
- Israel Ramos Villullas
- Merck Sharp & Dohme Research Laboratories, Neuroscience Research Centre, Terlings Park, Harlow, Essex, United Kingdom
| | | | | | | |
Collapse
|
38
|
Abstract
Sphingosine-1-phosphate (SIP) is a bioactive sphingolipid metabolite that regulates diverse cellular responses including, growth, survival, cytoskeleton rearrangements and movement. SIP plays an important role during development, particularly in vascular maturation and has been implicated in pathophysiology of cancer, wound healing, and atherosclerosis. This review summarizes the evidence showing that signaling induced by SIP is complex and involves both intracellular and extracellular actions. The intracellular effects of SIP remain speculative awaiting the identification of specific targets whereas the extracellular effects of SIP are clearly mediated through the activation of five specific G protein coupled receptors, called S1P1-5. Recent studies demonstrate that intracellular generated SIP can act in a paracrine or autocrine manner to activate its cell surface receptors.
Collapse
Affiliation(s)
- Kenneth Watterson
- Department of Biochemistry, Medical College of Virginia Campus, Virginia Commonwealth University, Tichmond, VA 23298, USA
| | | | | | | |
Collapse
|
39
|
Tilly JL, Kolesnick RN. Sphingolipids, apoptosis, cancer treatments and the ovary: investigating a crime against female fertility. BIOCHIMICA ET BIOPHYSICA ACTA 2002; 1585:135-8. [PMID: 12531546 DOI: 10.1016/s1388-1981(02)00333-5] [Citation(s) in RCA: 63] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Premature ovarian failure and infertility are well-known side-effects observed in young girls and reproductive-age women treated for cancer. Although the need for tumor eradication in these patients is clear, the long-term consequences of chemotherapy and radiation on non-target tissues, such as the ovaries where large numbers of germ cells (oocytes) are also killed off, are substantial. Unfortunately, the mechanism mediating the undesirable toxicity of cancer therapies in the female gonads has only recently been explored. Nevertheless, some important insights into the role of ceramide and sphingosine-1-phosphate (S1P) as a mediator and suppressor, respectively, of cancer therapy-induced oocyte apoptosis have emerged over the past few years. Such findings are exciting in that a better understanding of the crime--how radiation and chemotherapy kill off this irreplaceable population of innocent cells in the ovaries--may finally allow for the development of novel lipid-based strategies to combat infertility and premature menopause in female cancer patients.
Collapse
Affiliation(s)
- Jonathan L Tilly
- Vincent Center for Reproductive Biology, Department of Obstetrics and Gynecology, Massachusetts General Hospital/Harvard Medical School, 55 Fruit Street, VBK137E-GYN, Boston, MA 02114-2696, USA.
| | | |
Collapse
|
40
|
Osada M, Yatomi Y, Ohmori T, Ikeda H, Ozaki Y. Enhancement of sphingosine 1-phosphate-induced migration of vascular endothelial cells and smooth muscle cells by an EDG-5 antagonist. Biochem Biophys Res Commun 2002; 299:483-7. [PMID: 12445827 DOI: 10.1016/s0006-291x(02)02671-2] [Citation(s) in RCA: 160] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Sphingosine 1-phosphate (Sph-1-P), a bioactive lysophospholipid capable of inducing a wide spectrum of biological responses, acts as an intercellular mediator, through interaction with the endothelial differentiation gene (EDG)/S1P family of G protein-coupled receptors. In this study, the effects of JTE-013, a specific antagonist of the migration-inhibitory receptor EDG-5, on Sph-1-P-elicited responses were examined in human umbilical vein endothelial cells (HUVECs) and vascular smooth muscle cells (SMCs), which expressed EDG-5 protein weakly and abundantly, respectively. This pyrazolopyridine compound reversed the inhibitory effect of Sph-1-P on SMC migration and further enhanced Sph-1-P-stimulated HUVEC migration. In contrast, its effect on Sph-1-P-induced intracellular Ca(2+) mobilization was marginal. Our results indicate that specific regulation of Sph-1-P-modulated migration responses in vascular cells can be achieved by EDG-5 antagonists and that manipulation of Sph-1-P biological activities by each EDG antagonist may lead to a therapeutical application to control vascular diseases.
Collapse
MESH Headings
- Calcium Signaling/physiology
- Cell Movement/drug effects
- Cell Movement/physiology
- Cells, Cultured
- Endothelium, Vascular/cytology
- Endothelium, Vascular/drug effects
- Endothelium, Vascular/metabolism
- Fluorescent Dyes/metabolism
- Fura-2/metabolism
- Humans
- Immediate-Early Proteins/metabolism
- Lysophospholipids
- Muscle, Smooth, Vascular/cytology
- Myocytes, Smooth Muscle/cytology
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/metabolism
- Pyrazoles/chemistry
- Pyridines/chemistry
- Receptors, Cell Surface/antagonists & inhibitors
- Receptors, G-Protein-Coupled
- Receptors, Lysophospholipid
- Sphingosine/analogs & derivatives
- Sphingosine/pharmacology
Collapse
Affiliation(s)
- Makoto Osada
- Department of Laboratory Medicine, Yamanashi Medical University, Nakakoma, Yamanashi, Japan
| | | | | | | | | |
Collapse
|
41
|
Wang L, Cummings R, Usatyuk P, Morris A, Irani K, Natarajan V. Involvement of phospholipases D1 and D2 in sphingosine 1-phosphate-induced ERK (extracellular-signal-regulated kinase) activation and interleukin-8 secretion in human bronchial epithelial cells. Biochem J 2002; 367:751-60. [PMID: 12149127 PMCID: PMC1222936 DOI: 10.1042/bj20020586] [Citation(s) in RCA: 56] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2002] [Revised: 07/29/2002] [Accepted: 07/30/2002] [Indexed: 11/17/2022]
Abstract
Sphingosine 1-phosphate (S1P), a metabolite of sphingomyelin degradation, stimulates interleukin-8 (IL-8) secretion in human bronchial epithelial (Beas-2B) cells. The molecular mechanisms regulating S1P-mediated IL-8 secretion are yet to be completely defined. Here we provide evidence that activation of phospholipases D1 and D2 (PLD1 and PLD2) by S1P regulates the phosphorylation of extracellular-signal-regulated kinase (ERK) and IL-8 secretion in Beas-2B cells. S1P, in a time- and dose-dependent manner, enhanced the threonine/tyrosine phosphorylation of ERK. The inhibition of S1P-induced ERK phosphorylation by pertussis toxin and PD 98059 indicated coupling of S1P receptors to G(i) and the ERK signalling cascade respectively. Treatment of Beas-2B cells with butan-1-ol, but not butan-3-ol, abrogated the S1P-induced phosphorylation of Raf-1 and ERK, suggesting that PLD is involved in this activation. The roles of PLD1 and PLD2 in ERK activation and IL-8 secretion activated by S1P were investigated by infecting cells with adenoviral constructs of wild-type and catalytically inactive mutants of PLD1 and PLD2. Infection of Beas-2B cells with the wild-type constructs resulted in the activation of PLD1 and PLD2 by S1P and PMA. Also, the enhanced production of [(32)P]phosphatidic acid and [(32)P]phosphatidylbutanol in the presence of butan-1-ol and the increased phosphorylation of ERK by S1P were blocked by the catalytically inactive mutants hPLD1-K898R and mPLD2-K758R. Transient transfection of Beas-2B cells with human PLD1 and mouse PLD2 cDNAs potentiated S1P-mediated IL-8 secretion compared with vector controls. In addition, PD 98059 attenuated IL-8 secretion induced by S1P in a dose-dependent fashion. These results demonstrate that both PLD1 and PLD2 participate in S1P stimulation of ERK phosphorylation and IL-8 secretion in bronchial epithelial cells.
Collapse
Affiliation(s)
- Lixin Wang
- Department of Medicine, Division of Pulmonary and Critical Care, Johns Hopkins University, Baltimore, MD 21224, USA
| | | | | | | | | | | |
Collapse
|
42
|
Osada M, Yatomi Y, Ohmori T, Hosogaya S, Ozaki Y. Modulation of sphingosine 1-phosphate/EDG signaling by tumor necrosis factor-alpha in vascular endothelial cells. Thromb Res 2002; 108:169-74. [PMID: 12590954 DOI: 10.1016/s0049-3848(02)00385-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Affiliation(s)
- Makoto Osada
- Department of Clinical Laboratory, Yamanashi Medical University Hospital, Nakakoma, Japan
| | | | | | | | | |
Collapse
|
43
|
Ishii I, Ye X, Friedman B, Kawamura S, Contos JJA, Kingsbury MA, Yang AH, Zhang G, Brown JH, Chun J. Marked perinatal lethality and cellular signaling deficits in mice null for the two sphingosine 1-phosphate (S1P) receptors, S1P(2)/LP(B2)/EDG-5 and S1P(3)/LP(B3)/EDG-3. J Biol Chem 2002; 277:25152-9. [PMID: 12006579 DOI: 10.1074/jbc.m200137200] [Citation(s) in RCA: 203] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Five cognate G protein-coupled receptors (S1P(1-5)) have been shown to mediate various cellular effects of sphingosine 1-phosphate (S1P). Here we report the generation of mice null for S1P(2) and for both S1P(2) and S1P(3). S1P(2)-null mice were viable and fertile and developed normally. The litter sizes from S1P(2)S1P(3) double-null crosses were remarkably reduced compared with controls, and double-null pups often did not survive through infancy, although double-null survivors lacked any obvious phenotype. Mouse embryonic fibroblasts (MEFs) were examined for the effects of receptor deletions on S1P signaling pathways. Wild-type MEFs were responsive to S1P in activation of Rho and phospholipase C (PLC), intracellular calcium mobilization, and inhibition of forskolin-activated adenylyl cyclase. S1P(2)-null MEFs showed a significant decrease in Rho activation, but no effect on PLC activation, calcium mobilization, or adenylyl cyclase inhibition. Double-null MEFs displayed a complete loss of Rho activation and a significant decrease in PLC activation and calcium mobilization, with no effect on adenylyl cyclase inhibition. These data extend our previous findings on S1P(3)-null mice and indicate preferential coupling of the S1P(2) and S1P(3) receptors to Rho and PLC/Ca(2+) pathways, respectively. Although either receptor subtype supports embryonic development, deletion of both produces marked perinatal lethality, demonstrating an essential role for combined S1P signaling by these receptors.
Collapse
Affiliation(s)
- Isao Ishii
- Department of Pharmacology, School of Medicine, University of California at San Diego, La Jolla 92093-0636, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Ryu Y, Takuwa N, Sugimoto N, Sakurada S, Usui S, Okamoto H, Matsui O, Takuwa Y. Sphingosine-1-phosphate, a platelet-derived lysophospholipid mediator, negatively regulates cellular Rac activity and cell migration in vascular smooth muscle cells. Circ Res 2002; 90:325-32. [PMID: 11861422 DOI: 10.1161/hh0302.104455] [Citation(s) in RCA: 165] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Previous studies demonstrated that sphingosine-1-phosphate (S1P) induced migration of human umbilical vein endothelial cells (HUVECs) whereas it inhibited that of vascular smooth muscle cells (SMCs). This study explored the molecular mechanisms underlying the contrasting S1P actions on vascular cell motility. In rat and human aortic SMCs, the chemoattractant platelet-derived growth factor B-chain (PDGF) induced rapid 5- to 6-fold increases in the cellular amount of GTP-bound, active form of Rac. S1P did not affect PDGF-stimulated tyrosine phosphorylation of PDGF-beta receptor, but strongly inhibited PDGF-induced Rac activation, with a dose-response relationship similar to that for inhibition of PDGF-elicited chemotaxis. Dihydrosphingosine-1-phosphate, which is a weaker agonist for the S1P receptors, but not an inactive ligand sphingosine, also inhibited PDGF-stimulated chemotaxis and Rac activation although to lesser extents compared with S1P, suggesting that negative regulation by S1P of both chemotaxis and Rac was a receptor-mediated process. In contrast, S1P by itself stimulated Rac activity in HUVECs. Among the five S1P receptor isoforms, SMCs prominently expressed Edg-5 mRNA, whereas HUVECs expressed abundant Edg-1 mRNA but lacked detectable expression of Edg-5 mRNA. Adenovirus-mediated expression of a dominant-negative form of either Rac or Cdc42, but not RhoA, markedly attenuated chemotaxis of SMCs and HUVECs toward PDGF and S1P, respectively. Overexpression of Edg-1 in SMCs and Edg-5 in HUVECs reduced S1P-induced inhibition and stimulation, respectively, of Rac activity and migration. These results together indicate that Edg isoform-specific, negative or positive regulation of cellular Rac activity is critically involved in S1P-mediated bimodal regulation of cell motility in SMCs and HUVECs.
Collapse
MESH Headings
- Animals
- Blood Platelets/metabolism
- Cell Movement/drug effects
- Cell Movement/physiology
- Cell Surface Extensions/drug effects
- Cells, Cultured
- Chemotaxis/drug effects
- Chemotaxis/physiology
- Dose-Response Relationship, Drug
- Endothelium, Vascular/cytology
- Endothelium, Vascular/drug effects
- Endothelium, Vascular/metabolism
- Enzyme Activation/drug effects
- Humans
- Immediate-Early Proteins/biosynthesis
- Immediate-Early Proteins/genetics
- Immediate-Early Proteins/pharmacology
- Lysophospholipids/metabolism
- Lysophospholipids/pharmacology
- Muscle, Smooth, Vascular/cytology
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/metabolism
- Nuclear Proteins/biosynthesis
- Nuclear Proteins/genetics
- Nuclear Proteins/pharmacology
- Platelet-Derived Growth Factor/antagonists & inhibitors
- Platelet-Derived Growth Factor/pharmacology
- Rats
- Receptor, Platelet-Derived Growth Factor beta/metabolism
- Receptors, Cell Surface
- Receptors, G-Protein-Coupled
- Receptors, Lysophosphatidic Acid
- Receptors, Lysophospholipid
- Sphingosine/analogs & derivatives
- Sphingosine/metabolism
- Sphingosine/pharmacology
- Transcription Factors/biosynthesis
- Transcription Factors/genetics
- Transcription Factors/pharmacology
- cdc42 GTP-Binding Protein/genetics
- cdc42 GTP-Binding Protein/metabolism
- rac GTP-Binding Proteins/drug effects
- rac GTP-Binding Proteins/genetics
- rac GTP-Binding Proteins/metabolism
- rhoA GTP-Binding Protein/genetics
- rhoA GTP-Binding Protein/metabolism
Collapse
Affiliation(s)
- Yasuji Ryu
- Department of Physiology, Kanazawa University, Graduate School of Medicine, Kanazawa, Ishikawa, Japan
| | | | | | | | | | | | | | | |
Collapse
|
45
|
Banno Y, Takuwa Y, Akao Y, Okamoto H, Osawa Y, Naganawa T, Nakashima S, Suh PG, Nozawa Y. Involvement of phospholipase D in sphingosine 1-phosphate-induced activation of phosphatidylinositol 3-kinase and Akt in Chinese hamster ovary cells overexpressing EDG3. J Biol Chem 2001; 276:35622-8. [PMID: 11468290 DOI: 10.1074/jbc.m105673200] [Citation(s) in RCA: 56] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Phospholipase D (PLD), phosphatidylinositol 3-kinase (PI3K), and Akt are known to be involved in cellular signaling related to proliferation and cell survival. In this report, we provide evidence that PLD links sphingosine 1-phosphate (S1P)-induced activation of the G protein-coupled EDG3 receptor to stimulation of PI3K and its downstream effector Akt in Chinese hamster ovary (CHO) cells. S1P stimulation of EDG3-overexpressing CHO cells but not vector-transfected cells induced activation of PLD, PI3K, and Akt in a time- and dose-dependent manner. Akt phosphorylation was prevented by the PI3K inhibitors wortmannin and LY294002 (2-(4-monrpholinyl)-8-phenyl-4H-1-benzopyran-4-one), indicating that Akt activation was dependent on PI3K. S1P-induced activation of PI3K and Akt was abrogated by 1-butanol, which inhibited S1P-induced accumulation of phosphatidic acid by serving as a phosphatidyl group acceptor in the transphosphatidylation reaction catalyzed by PLD, whereas both PI3K and Akt activation were not inhibited by 2-butanol without such reaction. Co-expression of wild-type PLD2 with myc-Akt resulted in increased Akt activation in response to S1P. In contrast, co-expression of a catalytically inactive mutant of PLD2 eliminated the S1P-induced Akt activation. The treatment of EDG3-expressing CHO cells with exogenous Streptomyces chromofuscus PLD, which caused an accumulation of phosphatidic acid, resulted in increases in PI3K activity and the phosphorylation of Akt, the latter of which was completely abolished by LY294002. Furthermore, S1P-induced membrane ruffling, which was dependent on PI3K and Rac, was inhibited by 1-butanol, but not by 2-butanol. These results demonstrate that PLD participates in the activation of PI3K and Akt stimulation of EDG3 receptor.
Collapse
Affiliation(s)
- Y Banno
- Departments of Biochemistry and Internal Medicine, Gifu University School of Medicine, Gifu 500-8705, Japan.
| | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Okamoto H, Takuwa N, Yokomizo T, Sugimoto N, Sakurada S, Shigematsu H, Takuwa Y. Inhibitory regulation of Rac activation, membrane ruffling, and cell migration by the G protein-coupled sphingosine-1-phosphate receptor EDG5 but not EDG1 or EDG3. Mol Cell Biol 2000; 20:9247-61. [PMID: 11094076 PMCID: PMC102182 DOI: 10.1128/mcb.20.24.9247-9261.2000] [Citation(s) in RCA: 257] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Sphingosine-1-phosphate (S1P) is a bioactive lysophospholipid that induces a variety of biological responses in diverse cell types. Many, if not all, of these responses are mediated by members of the EDG (endothelial differentiation gene) family G protein-coupled receptors EDG1, EDG3, and EDG5 (AGR16). Among prominent activities of S1P is the regulation of cell motility; S1P stimulates or inhibits cell motility depending on cell types. In the present study, we provide evidence for EDG subtype-specific, contrasting regulation of cell motility and cellular Rac activity. In CHO cells expressing EDG1 or EDG3 (EDG1 cells or EDG3 cells, respectively) S1P as well as insulin-like growth factor I (IGF I) induced chemotaxis and membrane ruffling in phosphoinositide (PI) 3-kinase- and Rac-dependent manners. Both S1P and IGF I induced a biphasic increase in the amount of the GTP-bound active form of Rac. In CHO cells expressing EDG5 (EDG5 cells), IGF I similarly stimulated cell migration; however, in contrast to what was found for EDG1 and EDG3 cells, S1P did not stimulate migration but totally abolished IGF I-directed chemotaxis and membrane ruffling, in a manner dependent on a concentration gradient of S1P. In EDG5 cells, S1P stimulated PI 3-kinase activity as it did in EDG1 cells but inhibited the basal Rac activity and totally abolished IGF I-induced Rac activation, which involved stimulation of Rac-GTPase-activating protein activity rather than inhibition of Rac-guanine nucleotide exchange activity. S1P induced comparable increases in the amounts of GTP-RhoA in EDG3 and EDG5 cells. Neither S1P nor IGF I increased the amount of GTP-bound Cdc42. However, expression of N(17)-Cdc42, but not N(19)-RhoA, suppressed S1P- and IGF I-directed chemotaxis, suggesting a requirement for basal Cdc42 activity for chemotaxis. Taken together, the present results demonstrate that EDG5 is the first example of a hitherto-unrecognized type of receptors that negatively regulate Rac activity, thereby inhibiting cell migration and membrane ruffling.
Collapse
Affiliation(s)
- H Okamoto
- Department of Physiology, Kanazawa University School of Medicine, Kanazawa, Japan
| | | | | | | | | | | | | |
Collapse
|