1
|
Duse DA, Schröder NH, Srivastava T, Benkhoff M, Vogt J, Nowak MK, Funk F, Semleit N, Wollnitzke P, Erkens R, Kötter S, Meuth SG, Keul P, Santos W, Polzin A, Kelm M, Krüger M, Schmitt J, Levkau B. Deficiency of the sphingosine-1-phosphate (S1P) transporter Mfsd2b protects the heart against hypertension-induced cardiac remodeling by suppressing the L-type-Ca 2+ channel. Basic Res Cardiol 2024; 119:853-868. [PMID: 39110173 PMCID: PMC11461684 DOI: 10.1007/s00395-024-01073-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Revised: 07/26/2024] [Accepted: 07/29/2024] [Indexed: 10/09/2024]
Abstract
The erythrocyte S1P transporter Mfsd2b is also expressed in the heart. We hypothesized that S1P transport by Mfsd2b is involved in cardiac function. Hypertension-induced cardiac remodeling was induced by 4-weeks Angiotensin II (AngII) administration and assessed by echocardiography. Ca2+ transients and sarcomere shortening were examined in adult cardiomyocytes (ACM) from Mfsd2b+/+ and Mfsd2b-/- mice. Tension and force development were measured in skinned cardiac fibers. Myocardial gene expression was determined by real-time PCR, Protein Phosphatase 2A (PP2A) by enzymatic assay, and S1P by LC/MS, respectively. Msfd2b was expressed in the murine and human heart, and its deficiency led to higher cardiac S1P. Mfsd2b-/- mice had regular basal cardiac function but were protected against AngII-induced deterioration of left-ventricular function as evidenced by ~ 30% better stroke volume and cardiac index, and preserved ejection fraction despite similar increases in blood pressure. Mfsd2b-/- ACM exhibited attenuated Ca2+ mobilization in response to isoprenaline whereas contractility was unchanged. Mfsd2b-/- ACM showed no changes in proteins responsible for Ca2+ homeostasis, and skinned cardiac fibers exhibited reduced passive tension generation with preserved contractility. Verapamil abolished the differences in Ca2+ mobilization between Mfsd2b+/+ and Mfsd2b-/- ACM suggesting that S1P inhibits L-type-Ca2+ channels (LTCC). In agreement, intracellular S1P activated the inhibitory LTCC phosphatase PP2A in ACM and PP2A activity was increased in Mfsd2b-/- hearts. We suggest that myocardial S1P protects from hypertension-induced left-ventricular remodeling by inhibiting LTCC through PP2A activation. Pharmacologic inhibition of Mfsd2b may thus offer a novel approach to heart failure.
Collapse
Affiliation(s)
- Dragos Andrei Duse
- Institute for Molecular Medicine III, University Hospital Düsseldorf and Heinrich Heine University, Düsseldorf, Germany
- Department of Cardiology, Pneumology, and Vascular Medicine, Medical Faculty, Heinrich Heine University, Düsseldorf, Germany
- Cardiovascular Research Institute Düsseldorf (CARID), Düsseldorf, Germany
| | - Nathalie Hannelore Schröder
- Institute for Molecular Medicine III, University Hospital Düsseldorf and Heinrich Heine University, Düsseldorf, Germany
| | - Tanu Srivastava
- Institute of Pharmacology, University Hospital Düsseldorf, Düsseldorf, Germany
| | - Marcel Benkhoff
- Department of Cardiology, Pneumology, and Vascular Medicine, Medical Faculty, Heinrich Heine University, Düsseldorf, Germany
| | - Jens Vogt
- Institute for Molecular Medicine III, University Hospital Düsseldorf and Heinrich Heine University, Düsseldorf, Germany
| | - Melissa Kim Nowak
- Institute for Molecular Medicine III, University Hospital Düsseldorf and Heinrich Heine University, Düsseldorf, Germany
| | - Florian Funk
- Institute of Pharmacology, University Hospital Düsseldorf, Düsseldorf, Germany
| | - Nina Semleit
- Institute for Molecular Medicine III, University Hospital Düsseldorf and Heinrich Heine University, Düsseldorf, Germany
| | - Philipp Wollnitzke
- Institute for Molecular Medicine III, University Hospital Düsseldorf and Heinrich Heine University, Düsseldorf, Germany
| | - Ralf Erkens
- Department of Cardiology, Pneumology, and Vascular Medicine, Medical Faculty, Heinrich Heine University, Düsseldorf, Germany
- Cardiovascular Research Institute Düsseldorf (CARID), Düsseldorf, Germany
| | - Sebastian Kötter
- Institute of Cardiovascular Physiology, Medical Faculty and University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - Sven Günther Meuth
- Department of Neurology, Medical Faculty, Heinrich Heine University of Düsseldorf, Düsseldorf, Germany
| | - Petra Keul
- Institute for Molecular Medicine III, University Hospital Düsseldorf and Heinrich Heine University, Düsseldorf, Germany
| | - Webster Santos
- Department of Chemistry and Virginia Tech Center for Drug Discovery, Virginia Tech, Blacksburg, VA, 24060, USA
| | - Amin Polzin
- Department of Cardiology, Pneumology, and Vascular Medicine, Medical Faculty, Heinrich Heine University, Düsseldorf, Germany
- Cardiovascular Research Institute Düsseldorf (CARID), Düsseldorf, Germany
| | - Malte Kelm
- Department of Cardiology, Pneumology, and Vascular Medicine, Medical Faculty, Heinrich Heine University, Düsseldorf, Germany
- Cardiovascular Research Institute Düsseldorf (CARID), Düsseldorf, Germany
| | - Martina Krüger
- Cardiovascular Research Institute Düsseldorf (CARID), Düsseldorf, Germany
- Institute of Cardiovascular Physiology, Medical Faculty and University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - Joachim Schmitt
- Cardiovascular Research Institute Düsseldorf (CARID), Düsseldorf, Germany
- Institute of Pharmacology, University Hospital Düsseldorf, Düsseldorf, Germany
| | - Bodo Levkau
- Institute for Molecular Medicine III, University Hospital Düsseldorf and Heinrich Heine University, Düsseldorf, Germany.
- Cardiovascular Research Institute Düsseldorf (CARID), Düsseldorf, Germany.
| |
Collapse
|
2
|
Petrusca DN, Lee KP, Galson DL. Role of Sphingolipids in Multiple Myeloma Progression, Drug Resistance, and Their Potential as Therapeutic Targets. Front Oncol 2022; 12:925807. [PMID: 35756630 PMCID: PMC9213658 DOI: 10.3389/fonc.2022.925807] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Accepted: 05/16/2022] [Indexed: 11/13/2022] Open
Abstract
Multiple myeloma (MM) is an incapacitating hematological malignancy characterized by accumulation of cancerous plasma cells in the bone marrow (BM) and production of an abnormal monoclonal protein (M-protein). The BM microenvironment has a key role in myeloma development by facilitating the growth of the aberrant plasma cells, which eventually interfere with the homeostasis of the bone cells, exacerbating osteolysis and inhibiting osteoblast differentiation. Recent recognition that metabolic reprograming has a major role in tumor growth and adaptation to specific changes in the microenvironmental niche have led to consideration of the role of sphingolipids and the enzymes that control their biosynthesis and degradation as critical mediators of cancer since these bioactive lipids have been directly linked to the control of cell growth, proliferation, and apoptosis, among other cellular functions. In this review, we present the recent progress of the research investigating the biological implications of sphingolipid metabolism alterations in the regulation of myeloma development and its progression from the pre-malignant stage and discuss the roles of sphingolipids in in MM migration and adhesion, survival and proliferation, as well as angiogenesis and invasion. We introduce the current knowledge regarding the role of sphingolipids as mediators of the immune response and drug-resistance in MM and tackle the new developments suggesting the manipulation of the sphingolipid network as a novel therapeutic direction for MM.
Collapse
Affiliation(s)
- Daniela N Petrusca
- Department of Medicine, Division of Hematology/Oncology, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Kelvin P Lee
- Department of Medicine, Division of Hematology/Oncology, Indiana University School of Medicine, Indianapolis, IN, United States.,Indiana University Melvin and Bren Simon Comprehensive Cancer Center, Indianapolis, IN, United States
| | - Deborah L Galson
- Department of Medicine, Division of Hematology/Oncology, University of Pittsburgh Medical Center (UPMC) Hillman Cancer Center, McGowan Institute for Regenerative Medicine, HCC Research Pavilion, University of Pittsburgh, Pittsburgh, PA, United States
| |
Collapse
|
3
|
Mork BE, Lamerand SR, Zhou S, Taylor BK, Sheets PL. Sphingosine-1-phosphate receptor 1 agonist SEW2871 alters membrane properties of late-firing somatostatin expressing neurons in the central lateral amygdala. Neuropharmacology 2022; 203:108885. [PMID: 34798130 PMCID: PMC8672675 DOI: 10.1016/j.neuropharm.2021.108885] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Revised: 11/10/2021] [Accepted: 11/12/2021] [Indexed: 11/29/2022]
Abstract
Sphingosine-1-phosphate (S1P) is a bioactive sphingolipid that mediates a wide spectrum of biological processes including apoptosis, immune response and inflammation. Here, we sought to understand how S1P signaling affects neuronal excitability in the central amygdala (CeA), which is a brain region associated with fear learning, aversive memory, and the affective dimension of pain. Because the G-protein coupled S1P receptor 1 (S1PR1) has been shown to be the primary mediator of S1P signaling, we utilized S1PR1 agonist SEW2871 and S1PR1 antagonist NIBR to determine a potential role of S1PR1 in altering the cellular physiology of neurons in the lateral division of the CeA (CeL) that share the neuronal lineage marker somatostatin (Sst). CeL-Sst neurons play a critical role in expression of conditioned fear and pain modulation. Here we used transgenic breeding strategies to identify fluorescently labeled CeL-Sst neurons for electrophysiological recordings. Using principal component analysis, we identified two primary subtypes of Sst neurons within the CeL in both male and female mice. We denoted the two types regular-firing (type A) and late-firing (type B) CeL-Sst neurons. In response to SEW2871 application, Type A neurons exhibited increased input resistance, while type B neurons displayed a depolarized resting membrane potential and voltage threshold, increased current threshold, and decreased voltage height. NIBR application had no effect on CeL Sst neurons, indicating the absence of tonic S1P-induced S1PR1. Our findings reveal subtypes of Sst neurons within the CeL that are uniquely affected by S1PR1 activation, which may have implications for how S1P alters supraspinal circuits.
Collapse
Affiliation(s)
- Briana E Mork
- Medical Neurosciences Graduate Program, USA; Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Sydney R Lamerand
- Department of Anesthesiology and Perioperative Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213, USA
| | - Shudi Zhou
- Medical Neurosciences Graduate Program, USA; Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Bradley K Taylor
- Department of Anesthesiology and Perioperative Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213, USA
| | - Patrick L Sheets
- Medical Neurosciences Graduate Program, USA; Department of Pharmacology and Toxicology, USA; Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, 46202, USA.
| |
Collapse
|
4
|
Biringer RG. Endocannabinoid signaling pathways: beyond CB1R and CB2R. J Cell Commun Signal 2021; 15:335-360. [PMID: 33978927 PMCID: PMC8222499 DOI: 10.1007/s12079-021-00622-6] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Accepted: 04/27/2021] [Indexed: 12/15/2022] Open
Abstract
The search for cannabinoid receptors other than CB1R and CB2R has been ongoing for over a decade. A number of orphan receptors have been proposed as potential cannabinoid receptors primarily based on phylogenic arguments and reactivity towards known endocannabinoids and phytocannabinoids. Seven putative cannabinoid receptors are described and discussed, and evidence for and against their inclusion in this category are presented.
Collapse
Affiliation(s)
- Roger Gregory Biringer
- College of Osteopathic Medicine, Lake Erie College of Osteopathic Medicine, Bradenton, FL, 34211, USA.
| |
Collapse
|
5
|
Venkatraman G, Tang X, Du G, Parisentti AM, Hemmings DG, Brindley DN. Lysophosphatidate Promotes Sphingosine 1-Phosphate Metabolism and Signaling: Implications for Breast Cancer and Doxorubicin Resistance. Cell Biochem Biophys 2021; 79:531-545. [PMID: 34415509 PMCID: PMC11948428 DOI: 10.1007/s12013-021-01024-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Accepted: 07/09/2021] [Indexed: 10/20/2022]
Abstract
Lysophosphatidate (LPA) and sphingosine 1-phosphate (S1P) promote vasculogenesis, angiogenesis, and wound healing by activating a plethora of overlapping signaling pathways that stimulate mitogenesis, cell survival, and migration. As such, maladaptive signaling by LPA and S1P have major effects in increasing tumor progression and producing poor patient outcomes after chemotherapy and radiotherapy. Many signaling actions of S1P and LPA are not redundant; each are vital in normal physiology and their metabolisms differ. In the present work, we studied how LPA signaling impacts S1P metabolism and signaling in MDA-MB-231 and MCF-7 breast cancer cells. LPA increased sphingosine kinase-1 (SphK1) synthesis and rapidly activated cytosolic SphK1 through association with membranes. Blocking phospholipase D activity attenuated the LPA-induced activation of SphK1 and the synthesis of ABCC1 and ABCG2 transporters that secrete S1P from cells. This effect was magnified in doxorubicin-resistant MCF-7 cells. LPA also facilitated S1P signaling by increasing mRNA expression for S1P1 receptors. Doxorubicin-resistant MCF-7 cells had increased S1P2 and S1P3 receptor expression and show increased LPA-induced SphK1 activation, increased expression of ABCC1, ABCG2 and greater S1P secretion. Thus, LPA itself and LPA-induced S1P signaling counteract doxorubicin-induced death of MCF-7 cells. We conclude from the present and previous studies that LPA promotes S1P metabolism and signaling to coordinately increase tumor growth and metastasis and decrease the effectiveness of chemotherapy and radiotherapy for breast cancer treatment.
Collapse
Affiliation(s)
- Ganesh Venkatraman
- Department of Biochemistry, University of Alberta, Edmonton, AB, T6G 2S2, Canada
| | - Xiaoyun Tang
- Department of Biochemistry, University of Alberta, Edmonton, AB, T6G 2S2, Canada
- Cancer Research Institute of Northern Alberta, University of Alberta, Edmonton, AB, T6G 2S2, Canada
| | - Guangwei Du
- Department of Integrative Biology & Pharmacology, University of Texas Health Science at Houston, Houston, TX, 77030, USA
| | - Amadeo M Parisentti
- Northern Ontario School of Medicine, Health Sciences North Research Institute, Sudbury, ON, P3E 2H2, Canada
| | - Denise G Hemmings
- Cancer Research Institute of Northern Alberta, University of Alberta, Edmonton, AB, T6G 2S2, Canada.
- Medical Microbiology and Immunology, Obstetrics and Gynecology, Women and Children's Health Research Institute, Li Ka Shing Institute of Virology, Cardiovascular Research Center, University of Alberta, Edmonton, AB, T6G 2S2, Canada.
| | - David N Brindley
- Department of Biochemistry, University of Alberta, Edmonton, AB, T6G 2S2, Canada.
- Cancer Research Institute of Northern Alberta, University of Alberta, Edmonton, AB, T6G 2S2, Canada.
| |
Collapse
|
6
|
Jendzjowsky NG, Roy A, Iftinca M, Barioni NO, Kelly MM, Herrington BA, Visser F, Altier C, Wilson RJA. PKCε stimulation of TRPV1 orchestrates carotid body responses to asthmakines. J Physiol 2020; 599:1335-1354. [PMID: 33180962 PMCID: PMC7898719 DOI: 10.1113/jp280749] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Accepted: 11/05/2020] [Indexed: 12/11/2022] Open
Abstract
Key points We have previously shown that carotid body stimulation by lysophosphatidic acid elicits a reflex stimulation of vagal efferent activity sufficient to cause bronchoconstriction in asthmatic rats. Here, we show that pathophysiological concentrations of asthma‐associated prototypical Th2 cytokines also stimulate the carotid bodies. Stimulation of the carotid bodies by these asthmakines involves a PKCε–transient receptor potential vanilloid 1 (TRPV1) signalling mechanism likely dependent on TRPV1 S502 and T704 phosphorylation sites. As the carotid bodies’ oxygen sensitivity is independent of PKCε–TRPV1 signalling, systemic blockade of PKCε may provide a novel therapeutic target to reduce allergen‐induced asthmatic bronchoconstriction. Consistent with the therapeutic potential of blocking the PKCε–TRPV1 pathway, systemic delivery of a PKCε‐blocking peptide suppresses asthmatic respiratory distress in response to allergen and reduces airway hyperresponsiveness to bradykinin.
Abstract The autonomic nervous system orchestrates organ‐specific, systemic and behavioural responses to inflammation. Recently, we demonstrated a vital role for lysophosphatidic acid in stimulating the primary autonomic oxygen chemoreceptors, the carotid bodies, in parasympathetic‐mediated asthmatic airway hyperresponsiveness. However, the cacophony of stimulatory factors and cellular mechanisms of carotid body activation are unknown. Therefore, we set out to determine the intracellular signalling involved in carotid body‐mediated sensing of asthmatic blood‐borne inflammatory mediators. We employed a range of in vitro and rat in situ preparations, site‐directed mutagenesis, patch‐clamp, nerve recordings and pharmacological inhibition to assess cellular signalling. We show that the carotid bodies are also sensitive to asthma‐associated prototypical Th2 cytokines which elicit sensory nerve excitation. This provides additional asthmatic ligands contributing to the previously established reflex arc resulting in efferent vagal activity and asthmatic bronchoconstriction. This novel sensing role for the carotid body is mediated by a PKCε‐dependent stimulation of transient receptor potential vanilloid 1 (TRPV1), likely via TRPV1 phosphorylation at sites T704 and S502. Importantly, carotid body oxygen sensing was unaffected by blocking either PKCε or TRPV1. Further, we demonstrate that systemic PKCε blockade reduces asthmatic respiratory distress in response to allergen and airway hyperresponsiveness. These discoveries support an inflammation‐dependent, oxygen‐independent function for the carotid body and suggest that targeting PKCε provides a novel therapeutic option to abate allergic airway disease without altering life‐saving autonomic hypoxic reflexes. We have previously shown that carotid body stimulation by lysophosphatidic acid elicits a reflex stimulation of vagal efferent activity sufficient to cause bronchoconstriction in asthmatic rats. Here, we show that pathophysiological concentrations of asthma‐associated prototypical Th2 cytokines also stimulate the carotid bodies. Stimulation of the carotid bodies by these asthmakines involves a PKCε–transient receptor potential vanilloid 1 (TRPV1) signalling mechanism likely dependent on TRPV1 S502 and T704 phosphorylation sites. As the carotid bodies’ oxygen sensitivity is independent of PKCε–TRPV1 signalling, systemic blockade of PKCε may provide a novel therapeutic target to reduce allergen‐induced asthmatic bronchoconstriction. Consistent with the therapeutic potential of blocking the PKCε–TRPV1 pathway, systemic delivery of a PKCε‐blocking peptide suppresses asthmatic respiratory distress in response to allergen and reduces airway hyperresponsiveness to bradykinin.
Collapse
Affiliation(s)
- Nicholas G Jendzjowsky
- The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, California, USA
| | - Arijit Roy
- Department of Physiology and Pharmacology, University of Calgary, Calgary, Alberta, Canada.,Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada.,Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Alberta, Canada
| | - Mircea Iftinca
- Department of Physiology and Pharmacology, University of Calgary, Calgary, Alberta, Canada.,Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada.,Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Nicole O Barioni
- Department of Physiology and Pharmacology, University of Calgary, Calgary, Alberta, Canada.,Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada.,Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Alberta, Canada
| | - Margaret M Kelly
- Department of Physiology and Pharmacology, University of Calgary, Calgary, Alberta, Canada.,Department of Pathology and Laboratory Medicine, University of Calgary, Calgary, Alberta, Canada.,Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Alberta, Canada.,Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Brittney A Herrington
- Department of Physiology and Pharmacology, University of Calgary, Calgary, Alberta, Canada.,Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada.,Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Alberta, Canada
| | - Frank Visser
- Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
| | - Christophe Altier
- Department of Physiology and Pharmacology, University of Calgary, Calgary, Alberta, Canada.,Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada.,Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Richard J A Wilson
- Department of Physiology and Pharmacology, University of Calgary, Calgary, Alberta, Canada.,Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada.,Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
7
|
Riboni L, Abdel Hadi L, Navone SE, Guarnaccia L, Campanella R, Marfia G. Sphingosine-1-Phosphate in the Tumor Microenvironment: A Signaling Hub Regulating Cancer Hallmarks. Cells 2020; 9:E337. [PMID: 32024090 PMCID: PMC7072483 DOI: 10.3390/cells9020337] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 01/27/2020] [Accepted: 01/29/2020] [Indexed: 02/07/2023] Open
Abstract
As a key hub of malignant properties, the cancer microenvironment plays a crucial role intimately connected to tumor properties. Accumulating evidence supports that the lysophospholipid sphingosine-1-phosphate acts as a key signal in the cancer extracellular milieu. In this review, we have a particular focus on glioblastoma, representative of a highly aggressive and deleterious neoplasm in humans. First, we highlight recent advances and emerging concepts for how tumor cells and different recruited normal cells contribute to the sphingosine-1-phosphate enrichment in the cancer microenvironment. Then, we describe and discuss how sphingosine-1-phosphate signaling contributes to favor cancer hallmarks including enhancement of proliferation, stemness, invasion, death resistance, angiogenesis, immune evasion and, possibly, aberrant metabolism. We also discuss the potential of how sphingosine-1-phosphate control mechanisms are coordinated across distinct cancer microenvironments. Further progress in understanding the role of S1P signaling in cancer will depend crucially on increasing knowledge of its participation in the tumor microenvironment.
Collapse
Affiliation(s)
- Laura Riboni
- Department of Medical Biotechnology and Translational Medicine, LITA-Segrate, University of Milan, via Fratelli Cervi, 93, 20090 Segrate, Milan, Italy
| | - Loubna Abdel Hadi
- Department of Medical Biotechnology and Translational Medicine, LITA-Segrate, University of Milan, via Fratelli Cervi, 93, 20090 Segrate, Milan, Italy
| | - Stefania Elena Navone
- Laboratory of Experimental Neurosurgery and Cell Therapy, Neurosurgery Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, via Francesco Sforza 35, 20122 Milan, Italy (L.G.)
| | - Laura Guarnaccia
- Laboratory of Experimental Neurosurgery and Cell Therapy, Neurosurgery Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, via Francesco Sforza 35, 20122 Milan, Italy (L.G.)
- Department of Clinical Sciences and Community Health, University of Milan, 20100 Milan, Italy
| | - Rolando Campanella
- Laboratory of Experimental Neurosurgery and Cell Therapy, Neurosurgery Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, via Francesco Sforza 35, 20122 Milan, Italy (L.G.)
| | - Giovanni Marfia
- Laboratory of Experimental Neurosurgery and Cell Therapy, Neurosurgery Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, via Francesco Sforza 35, 20122 Milan, Italy (L.G.)
| |
Collapse
|
8
|
Aktories K, Gierschik P, Heringdorf DMZ, Schmidt M, Schultz G, Wieland T. cAMP guided his way: a life for G protein-mediated signal transduction and molecular pharmacology-tribute to Karl H. Jakobs. Naunyn Schmiedebergs Arch Pharmacol 2019; 392:887-911. [PMID: 31101932 DOI: 10.1007/s00210-019-01650-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2019] [Accepted: 04/02/2019] [Indexed: 12/14/2022]
Abstract
Karl H. Jakobs, former editor-in-chief of Naunyn-Schmiedeberg's Archives of Pharmacology and renowned molecular pharmacologist, passed away in April 2018. In this article, his scientific achievements regarding G protein-mediated signal transduction and regulation of canonical pathways are summarized. Particularly, the discovery of inhibitory G proteins for adenylyl cyclase, methods for the analysis of receptor-G protein interactions, GTP supply by nucleoside diphosphate kinases, mechanisms in phospholipase C and phospholipase D activity regulation, as well as the development of the concept of sphingosine-1-phosphate as extra- and intracellular messenger will presented. His seminal scientific and methodological contributions are put in a general and timely perspective to display and honor his outstanding input to the current knowledge in molecular pharmacology.
Collapse
Affiliation(s)
- Klaus Aktories
- Institute for Experimental and Clinical Pharmacology and Toxicology, Faculty of Medicine, Albert Ludwigs University, 79104, Freiburg, Germany
| | - Peter Gierschik
- Institute of Pharmacology and Toxicology, Ulm University Medical Center, 89070, Ulm, Germany
| | - Dagmar Meyer Zu Heringdorf
- Institute of General Pharmacology and Toxicology, University Hospital Frankfurt am Main, Goethe University, 60590, Frankfurt am Main, Germany
| | - Martina Schmidt
- Department of Molecular Pharmacology, University of Groningen, 9713AV, Groningen, The Netherlands
| | - Günter Schultz
- Department of Pharmacology, Charité University Medical Center Berlin, Campus Benjamin Franklin, 14195, Berlin, Germany
| | - Thomas Wieland
- Experimental Pharmacology Mannheim (EPM), European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, Ludolf-Krehl-Str. 13 - 17, 68167, Mannheim, Germany.
| |
Collapse
|
9
|
N-Cinnamoylanthranilates as human TRPA1 modulators: Structure-activity relationships and channel binding sites. Eur J Med Chem 2019; 170:141-156. [DOI: 10.1016/j.ejmech.2019.02.074] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Revised: 02/27/2019] [Accepted: 02/27/2019] [Indexed: 01/14/2023]
|
10
|
S1P 1 receptor phosphorylation, internalization, and interaction with Rab proteins: effects of sphingosine 1-phosphate, FTY720-P, phorbol esters, and paroxetine. Biosci Rep 2018; 38:BSR20181612. [PMID: 30366961 PMCID: PMC6294635 DOI: 10.1042/bsr20181612] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2018] [Revised: 10/19/2018] [Accepted: 10/26/2018] [Indexed: 01/04/2023] Open
Abstract
Sphingosine 1-phosphate (S1P) and FTY720-phosphate (FTYp) increased intracellular calcium in cells expressing S1P1 mCherry-tagged receptors; the synthetic agonist was considerably less potent. Activation of protein kinase C by phorbol myristate acetate (PMA) blocked these effects. The three agents induced receptor phosphorylation and internalization, with the action of FTYp being more intense. S1P1 receptor–Rab protein (GFP-tagged) interaction was studied using FRET. The three agents were able to induce S1P1 receptor–Rab5 interaction, although with different time courses. S1P1 receptor–Rab9 interaction was mainly increased by the phorbol ester, whereas S1P1 receptor–Rab7 interaction was only increased by FTYp and after a 30-min incubation. These actions were not observed using dominant negative (GDP-bound) Rab protein mutants. The data suggested that the three agents induce interaction with early endosomes, but that the natural agonist induced rapid receptor recycling, whereas activation of protein kinase C favored interaction with late endosome and slow recycling and FTYp triggered receptor interaction with vesicles associated with proteasomal/lysosomal degradation. The ability of bisindolylmaleimide I and paroxetine to block some of these actions suggested the activation of protein kinase C was associated mainly with the action of PMA, whereas G protein-coupled receptor kinase (GRK) 2 (GRK2) was involved in the action of the three agents.
Collapse
|
11
|
Pierucci F, Frati A, Battistini C, Matteini F, Iachini MC, Vestri A, Penna F, Costelli P, Meacci E. Involvement of released sphingosine 1-phosphate/sphingosine 1-phosphate receptor axis in skeletal muscle atrophy. Biochim Biophys Acta Mol Basis Dis 2018; 1864:3598-3614. [PMID: 30279138 DOI: 10.1016/j.bbadis.2018.08.040] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2018] [Revised: 07/27/2018] [Accepted: 08/30/2018] [Indexed: 12/18/2022]
Abstract
Skeletal muscle (SkM) atrophy is caused by several and heterogeneous conditions, such as cancer, neuromuscular disorders and aging. In most types of SkM atrophy overall rates of protein synthesis are suppressed, protein degradation is consistently elevated and atrogenes, such as the ubiquitin ligase Atrogin-1/MAFbx, are up-regulated. The molecular regulators of SkM waste are multiple and only in part known. Sphingolipids represent a class of bioactive molecules capable of modulating the destiny of many cell types, including SkM cells. In particular, we and others have shown that sphingosine 1phosphate (S1P), formed by sphingosine kinase (SphK), is able to act as trophic and morphogenic factor in myoblasts. Here, we report the first evidence that the atrophic phenotype observed in both muscle obtained from mice bearing the C26 adenocarcinoma and C2C12 myotubes treated with dexamethasone was characterized by reduced levels of active phospho-SphK1. The importance of SphK1 activity is also confirmed by the specific pharmacological inhibition of SphK1 able to increase Atrogin-1/MAFbx expression and reduce myotube size and myonuclei number. Furthermore, we found that SkM atrophy was accomplished by significant increase of S1P transporter Spns2 and in changes in the pattern of S1P receptor (S1PRs) subtype expression paralleled by increased Atrogin-1/MAFbx expression, suggesting a role for the released S1P and of specific S1PR-mediated signaling pathways in the control of the ubiquitin ligase. Altogether, these findings provide the first evidence that SphK1/released S1P/S1PR axis acts as a molecular regulator of SkM atrophy, thereby representing a new possible target for therapy in many patho-physiological conditions.
Collapse
Affiliation(s)
- Federica Pierucci
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio" -Unit of Biochemical Sciences and Molecular Biology, University of Florence, Viale GB Morgagni, 50, Florence, Italy
| | - Alessia Frati
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio" -Unit of Biochemical Sciences and Molecular Biology, University of Florence, Viale GB Morgagni, 50, Florence, Italy
| | - Chiara Battistini
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio" -Unit of Biochemical Sciences and Molecular Biology, University of Florence, Viale GB Morgagni, 50, Florence, Italy
| | - Francesca Matteini
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio" -Unit of Biochemical Sciences and Molecular Biology, University of Florence, Viale GB Morgagni, 50, Florence, Italy
| | - Maria Chiara Iachini
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio" -Unit of Biochemical Sciences and Molecular Biology, University of Florence, Viale GB Morgagni, 50, Florence, Italy
| | - Ambra Vestri
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio" -Unit of Biochemical Sciences and Molecular Biology, University of Florence, Viale GB Morgagni, 50, Florence, Italy
| | - Fabio Penna
- Department of Clinical and Biological Sciences, University of Turin, Regione Gonzole 10, Orbassano (TO), Italy
| | - Paola Costelli
- Department of Clinical and Biological Sciences, University of Turin, Regione Gonzole 10, Orbassano (TO), Italy
| | - Elisabetta Meacci
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio" -Unit of Biochemical Sciences and Molecular Biology, University of Florence, Viale GB Morgagni, 50, Florence, Italy.
| |
Collapse
|
12
|
S1P Provokes Tumor Lymphangiogenesis via Macrophage-Derived Mediators Such as IL-1 β or Lipocalin-2. Mediators Inflamm 2017; 2017:7510496. [PMID: 28804221 PMCID: PMC5539930 DOI: 10.1155/2017/7510496] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2017] [Accepted: 06/15/2017] [Indexed: 12/17/2022] Open
Abstract
A pleiotropic signaling lipid, sphingosine-1-phosphate (S1P), has been implicated in various pathophysiological processes supporting tumor growth and metastasis. However, there are only a few descriptive studies suggesting a role of S1P in tumor lymphangiogenesis, which is critical for tumor growth and dissemination. Corroborating own data, the literature suggests that apoptotic tumor cell-derived S1P alters the phenotype of tumor-associated macrophages (TAMs) to gain protumor functions. However, mechanistically, the role of TAM-induced lymphangiogenesis has only been poorly described, mostly linked to the production of lymphangiogenic factors such as vascular endothelial growth factor C (VEGF-C) and VEGF-D, or transdifferentiation into lymphatic endothelial cells. Recent findings highlight a rather underappreciated role of S1P in tumor lymphangiogenesis, referring to the production of interleukin-1β (IL-1β) and lipocalin-2 (LCN2) by a tumor-promoting macrophage phenotype. In this review, we aim to provide to the readers with the current understanding of the molecular mechanism how apoptotic cell-derived S1P triggers TAMs to promote lymphangiogenesis.
Collapse
|
13
|
Dobierzewska A, Palominos M, Sanchez M, Dyhr M, Helgert K, Venegas-Araneda P, Tong S, Illanes SE. Impairment of Angiogenic Sphingosine Kinase-1/Sphingosine-1-Phosphate Receptors Pathway in Preeclampsia. PLoS One 2016; 11:e0157221. [PMID: 27284992 PMCID: PMC4902228 DOI: 10.1371/journal.pone.0157221] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2016] [Accepted: 05/26/2016] [Indexed: 12/17/2022] Open
Abstract
Preeclampsia (PE), is a serious pregnancy disorder characterized in the early gestation by shallow trophoblast invasion, impaired placental neo-angiogenesis, placental hypoxia and ischemia, which leads to maternal and fetal morbidity and mortality. Here we hypothesized that angiogenic sphingosine kinase-1 (SPHK1)/sphingosine-1-phosphate (S1P) receptors pathway is impaired in PE. We found that SPHK1 mRNA and protein expression are down-regulated in term placentae and term chorionic villous explants from patients with PE or severe PE (PES), compared with controls. Moreover, mRNA expression of angiogenic S1PR1 and S1PR3 receptors were decreased in placental samples of PE and PES patients, whereas anti-angiogenic S1PR2 was up-regulated in chorionic villous tissue of PES subjects, pointing to its potential atherogenic and inflammatory properties. Furthermore, in in vitro (JAR cells) and ex vivo (chorionic villous explants) models of placental hypoxia, SPHK1 mRNA and protein were strongly up-regulated under low oxygen tension (1% 02). In contrast, there was no change in SPHK1 expression under the conditions of placental physiological hypoxia (8% 02). In both models, nuclear protein levels of HIF1A were increased at 1% 02 during the time course, but there was no up-regulation at 8% 02, suggesting that SPHK1 and HIF1A might be the part of the same canonical pathway during hypoxia and that both contribute to placental neovascularization during early gestation. Taken together, this study suggest the SPHK1 pathway may play a role in the human early placentation process and may be involved in the pathogenesis of PE.
Collapse
Affiliation(s)
- Aneta Dobierzewska
- Department of Obstetrics & Gynecology and Laboratory of Reproductive Biology, Faculty of Medicine, Universidad de los Andes, Santiago, Chile
- * E-mail:
| | - Macarena Palominos
- Department of Obstetrics & Gynecology and Laboratory of Reproductive Biology, Faculty of Medicine, Universidad de los Andes, Santiago, Chile
| | - Marianela Sanchez
- Department of Obstetrics & Gynecology and Laboratory of Reproductive Biology, Faculty of Medicine, Universidad de los Andes, Santiago, Chile
| | - Michael Dyhr
- Department of Obstetrics & Gynecology and Laboratory of Reproductive Biology, Faculty of Medicine, Universidad de los Andes, Santiago, Chile
| | - Katja Helgert
- Department of Obstetrics & Gynecology and Laboratory of Reproductive Biology, Faculty of Medicine, Universidad de los Andes, Santiago, Chile
| | - Pia Venegas-Araneda
- Department of Obstetrics & Gynecology and Laboratory of Reproductive Biology, Faculty of Medicine, Universidad de los Andes, Santiago, Chile
| | - Stephen Tong
- Translational Obstetrics Group, Department of Obstetrics and Gynecology, University of Melbourne, Mercy Hospital for Women, Heidelberg, Victoria, Australia
| | - Sebastian E. Illanes
- Department of Obstetrics & Gynecology and Laboratory of Reproductive Biology, Faculty of Medicine, Universidad de los Andes, Santiago, Chile
- Department of Maternal-Fetal Medicine, Clinica Davila, Santiago, Chile
| |
Collapse
|
14
|
Sphingosine Kinases: Emerging Structure-Function Insights. Trends Biochem Sci 2016; 41:395-409. [PMID: 27021309 DOI: 10.1016/j.tibs.2016.02.007] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2015] [Revised: 02/08/2016] [Accepted: 02/17/2016] [Indexed: 12/15/2022]
Abstract
Sphingosine kinases (SK1 and SK2) catalyse the conversion of sphingosine into sphingosine 1-phosphate and control fundamental cellular processes, including cell survival, proliferation, differentiation, migration, and immune function. In this review, we highlight recent breakthroughs in the structural and functional characterisation of SK1 and these are contextualised by analysis of crystal structures for closely related prokaryotic lipid kinases. We identify a putative dimerisation interface and propose novel regulatory mechanisms governing structural plasticity induced by phosphorylation and interaction with phospholipids and proteins. Our analysis suggests that the catalytic function and regulation of the enzymes might be dependent on conformational mobility and it provides a roadmap for future interrogation of SK1 function and its role in physiology and disease.
Collapse
|
15
|
Pham DH, Powell JA, Gliddon BL, Moretti PAB, Tsykin A, Van der Hoek M, Kenyon R, Goodall GJ, Pitson SM. Enhanced expression of transferrin receptor 1 contributes to oncogenic signalling by sphingosine kinase 1. Oncogene 2013; 33:5559-68. [PMID: 24276247 DOI: 10.1038/onc.2013.502] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2013] [Revised: 10/10/2013] [Accepted: 10/19/2013] [Indexed: 12/25/2022]
Abstract
Sphingosine kinase 1 (SK1) is a lipid kinase that catalyses the formation of sphingosine-1-phosphate (S1P). Considerable evidence has implicated elevated cellular SK1 in tumour development, progression and disease severity. In particular, SK1 has been shown to enhance cell survival and proliferation and induce neoplastic transformation. Although S1P has been found to have both cell-surface G-protein-coupled receptors and intracellular targets, the specific downstream pathways mediating oncogenic signalling by SK1 remain poorly defined. Here, using a gene expression array approach, we have demonstrated a novel mechanism whereby SK1 regulates cell survival, proliferation and neoplastic transformation through enhancing expression of transferrin receptor 1 (TFR1). We showed that elevated levels of SK1 enhanced total as well as cell-surface TFR1 expression, resulting in increased transferrin uptake into cells. Notably, we also found that SK1 activation and localization to the plasma membrane, which are critical for its oncogenic effects, are necessary for regulation of TFR1 expression specifically through engagement of the S1P G-protein coupled receptor, S1P2. Furthermore, we showed that blocking TFR1 function with a neutralizing antibody inhibits SK1-induced cell proliferation, survival and neoplastic transformation of NIH3T3 fibroblasts. Similar effects were observed following antagonism of S1P2. Together these findings suggest that TFR1 has an important role in SK1-mediated oncogenesis.
Collapse
Affiliation(s)
- D H Pham
- 1] Centre for Cancer Biology, SA Pathology, Frome Road, Adelaide, SA, Australia [2] School of Molecular and Biomedical Science, University of Adelaide, Adelaide, SA, Australia
| | - J A Powell
- Centre for Cancer Biology, SA Pathology, Frome Road, Adelaide, SA, Australia
| | - B L Gliddon
- Centre for Cancer Biology, SA Pathology, Frome Road, Adelaide, SA, Australia
| | - P A B Moretti
- Centre for Cancer Biology, SA Pathology, Frome Road, Adelaide, SA, Australia
| | - A Tsykin
- 1] Centre for Cancer Biology, SA Pathology, Frome Road, Adelaide, SA, Australia [2] School of Molecular and Biomedical Science, University of Adelaide, Adelaide, SA, Australia
| | - M Van der Hoek
- Adelaide Microarray Facility, SA Pathology, Frome Road, Adelaide, SA, Australia
| | - R Kenyon
- Adelaide Microarray Facility, SA Pathology, Frome Road, Adelaide, SA, Australia
| | - G J Goodall
- 1] Centre for Cancer Biology, SA Pathology, Frome Road, Adelaide, SA, Australia [2] School of Molecular and Biomedical Science, University of Adelaide, Adelaide, SA, Australia [3] School of Pharmacy and Medical Science, University of South Australia, Adelaide, SA, Australia
| | - S M Pitson
- 1] Centre for Cancer Biology, SA Pathology, Frome Road, Adelaide, SA, Australia [2] School of Molecular and Biomedical Science, University of Adelaide, Adelaide, SA, Australia [3] School of Pharmacy and Medical Science, University of South Australia, Adelaide, SA, Australia
| |
Collapse
|
16
|
Selley DE, Welch SP, Sim-Selley LJ. Sphingosine lysolipids in the CNS: endogenous cannabinoid antagonists or a parallel pain modulatory system? Life Sci 2013; 93:187-93. [PMID: 23782998 DOI: 10.1016/j.lfs.2013.06.004] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2013] [Revised: 06/02/2013] [Accepted: 06/06/2013] [Indexed: 01/02/2023]
Abstract
A significant number of patients experience chronic pain and the intractable side effects of currently prescribed pain medications. Recent evidence indicates important pain-modulatory roles for two classes of G-protein-coupled receptors that are activated by endogenous lipid ligands, the endocannabinoid (eCB) and sphingosine-1-phosphate (S1P) receptors, which are widely expressed in both the immune and nervous systems. In the central nervous system (CNS), CB1 cannabinoid and S1P1 receptors are most abundantly expressed and exhibit overlapping anatomical distributions and similar signaling mechanisms. The eCB system has emerged as a potential target for treatment of chronic pain, but comparatively little is known about the roles of S1P in pain regulation. Both eCB and S1P systems modulate pain perception via the central and peripheral nervous systems. In most paradigms studied, the eCB system mainly inhibits pain perception. In contrast, S1P acting peripherally at S1P1 and S1P3 receptors can enhance sensitivity to various pain stimuli or elicit spontaneous pain. However, S1P acting at S1P1 receptors and possibly other targets in the CNS can attenuate sensitivity to various pain stimuli. Interestingly, other endogenous sphingolipid derivatives might play a role in central pain sensitization. Moreover, these sphingolipids can also act as CB1 cannabinoid receptor antagonists, but the physiological relevance of this interaction is unknown. Overall, both eCB and sphingolipid systems offer promising targets for the treatment of chronic pain. This review compares and contrasts the eCB and S1P systems with a focus on their roles in pain modulation, and considers possible points of interaction between these systems.
Collapse
Affiliation(s)
- Dana E Selley
- Department of Pharmacology and Toxicology and Institute for Drug and Alcohol Studies, Virginia Commonwealth University, Richmond, VA 23298, United States.
| | | | | |
Collapse
|
17
|
Alshaker H, Sauer L, Monteil D, Ottaviani S, Srivats S, Böhler T, Pchejetski D. Therapeutic potential of targeting SK1 in human cancers. Adv Cancer Res 2013; 117:143-200. [PMID: 23290780 DOI: 10.1016/b978-0-12-394274-6.00006-6] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Sphingosine kinase 1 (SK1) is a lipid enzyme with oncogenic properties that converts the proapoptotic lipids ceramide and sphingosine into the antiapoptotic lipid sphingosine-1-phosphate and activates the signal transduction pathways that lead to cell proliferation, migration, the activation of the inflammatory response, and the impairment of apoptosis. There is compelling evidence that SK1 activation contributes to cancer progression leading to increased oncogenic transformation, tumor growth, resistance to therapies, tumor neovascularization, and metastatic spread. High levels of SK1 expression or activity have been associated with a poor prognosis in several human cancers. Recent studies using cancer cell and mouse models demonstrate a significant potential for SK1-targeting therapies to synergize with the effects of chemotherapy and radiotherapy; however, until recently the absence of clinically applicable SK1 inhibitors has limited the translation of these findings into patients. With the recent discovery of SK1 inhibiting properties of a clinically approved drug FTY720 (Fingolimod), SK1 has gained significant attention from both clinicians and the pharmaceutical industry and it is hoped that trials of newly developed SK1 inhibitors may follow soon. This review provides an overview of the SK1 signaling, its relevance to cancer progression, and the potential clinical significance of targeting SK1 for improved local or systemic control of human cancers.
Collapse
Affiliation(s)
- Heba Alshaker
- Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, Hammersmith Hospital, London, United Kingdom
| | | | | | | | | | | | | |
Collapse
|
18
|
Kawabori M, Kacimi R, Karliner JS, Yenari MA. Sphingolipids in cardiovascular and cerebrovascular systems: Pathological implications and potential therapeutic targets. World J Cardiol 2013; 5:75-86. [PMID: 23675553 PMCID: PMC3653015 DOI: 10.4330/wjc.v5.i4.75] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2013] [Revised: 03/01/2013] [Accepted: 03/29/2013] [Indexed: 02/06/2023] Open
Abstract
The sphingolipid metabolites ceramide, sphingosine, and sphingosine-1-phosphate (S1P) and its enzyme sphingosine kinase (SphK) play an important role in the regulation of cell proliferation, survival, inflammation, and cell death. Ceramide and sphingosine usually inhibit proliferation and promote apoptosis, while its metabolite S1P phosphorylated by SphK stimulates growth and suppresses apoptosis. Because these metabolites are interconvertible, it has been proposed that it is not the absolute amounts of these metabolites but rather their relative levels that determine cell fate. The relevance of this “sphingolipid rheostat” and its role in regulating cell fate has been borne out by work in many labs using many different cell types and experimental manipulations. A central finding of these studies is that SphK is a critical regulator of the sphingolipid rheostat, as it not only produces the pro-growth, anti-apoptotic messenger S1P, but also decreases levels of pro-apoptotic ceramide and sphingosine. Activation of bioactive sphingolipid S1P signaling has emerged as a critical protective pathway in response to acute ischemic injury in both cardiac and cerebrovascular disease, and these observations have considerable relevance for future potential therapeutic targets.
Collapse
|
19
|
Post-translational regulation of sphingosine kinases. Biochim Biophys Acta Mol Cell Biol Lipids 2013; 1831:147-56. [DOI: 10.1016/j.bbalip.2012.07.005] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2012] [Revised: 07/04/2012] [Accepted: 07/06/2012] [Indexed: 12/22/2022]
|
20
|
Sphingosine-1-phosphate receptors as emerging targets for treatment of pain. Biochem Pharmacol 2012; 84:1551-62. [PMID: 22971335 DOI: 10.1016/j.bcp.2012.08.010] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2012] [Revised: 08/01/2012] [Accepted: 08/10/2012] [Indexed: 11/24/2022]
Abstract
Lysolipids are important mediators of cellular communication in multiple physiological processes. Sphingosine-1-phosphate (S1P) is a major lysolipid in many organs, including the central nervous system (CNS). This commentary discusses recent findings on the role of S1P in regulating pain perception, and highlights advances and challenges in the field. S1P interacts with multiple cellular targets, including G-protein-coupled receptors. Known S1P receptors include five types, four of which are expressed in the CNS (S1P(1,2,3,5)) where they are localized on neurons and glia. S1P receptor-mediated G-protein activation has been demonstrated throughout the CNS, including regions that regulate nociception. S1P receptors couple to multiple G-proteins to produce various intracellular responses, and can mediate both excitatory and inhibitory neuromodulation, depending on the receptor type and cellular context. Both antinociceptive and pro-nociceptive effects of S1P have been reported, and both actions can involve S1P(1) receptors. Current evidence suggests that antinociception is mediated by CNS neurons, whereas pro-nociception is mediated by primary afferent neurons or immune cells in the periphery, or CNS glia. Nonetheless, peripheral administration of the S1P(1,3,4,5) agonist pro-drug, FTY720, produces antinociception. FTY720 is approved to treat multiple sclerosis, and produces potent anti-inflammatory effects, which suggests potential utility for painful autoimmune diseases. Furthermore, evidence suggests that the S1P system interacts with other pain-modulatory systems, such as endogenous cannabinoid and opioid systems, and putative novel sphingolipid targets in the CNS. These findings suggest that drugs targeting the S1P system could be developed as novel analgesics, either as monotherapy or potential adjuncts to established analgesics.
Collapse
|
21
|
Antoon JW, White MD, Driver JL, Burow ME, Beckman BS. Sphingosine kinase isoforms as a therapeutic target in endocrine therapy resistant luminal and basal-A breast cancer. Exp Biol Med (Maywood) 2012; 237:832-44. [PMID: 22859737 DOI: 10.1258/ebm.2012.012028] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Sphingosine kinase signaling has become of increasing interest as a cancer target in recent years. Two sphingosine kinase inhibitors, sphingosine kinase inhibitor (SKI)-II and ABC294640, are promising as potential breast cancer therapies. However, evidence for their therapeutic properties in specific breast cancer subtypes is currently lacking. In this study, we characterize these drugs in luminal, endocrine-resistant (MDA-MB-361) and basal-A, triple-negative (MDA-MB-468) breast cancer cells and compare them with previously published data in other breast cancer cell models. Both SKI-II and ABC294640 demonstrated greater efficacy in basal-A compared with luminal breast cancer. ABC294640, in particular, induced apoptosis and blocked proliferation both in vitro and in vivo in this triple-negative breast cancer system. Furthermore, Sphk expression promotes survival and endocrine therapy resistance in previously sensitive breast cancer cells. Taken together, these results characterize sphingosine kinase inhibitors across breast cancer cell systems and demonstrate their therapeutic potential as anti-cancer agents.
Collapse
Affiliation(s)
- James W Antoon
- Tulane Department of Pharmacology, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | | | | | | | | |
Collapse
|
22
|
Liu X, Zhang QH, Yi GH. Regulation of metabolism and transport of sphingosine-1-phosphate in mammalian cells. Mol Cell Biochem 2011; 363:21-33. [DOI: 10.1007/s11010-011-1154-1] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2011] [Accepted: 11/11/2011] [Indexed: 02/04/2023]
|
23
|
Barra V, Kuhn AM, von Knethen A, Weigert A, Brüne B. Apoptotic cell-derived factors induce arginase II expression in murine macrophages by activating ERK5/CREB. Cell Mol Life Sci 2011; 68:1815-27. [PMID: 20949368 PMCID: PMC11115119 DOI: 10.1007/s00018-010-0537-x] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2010] [Revised: 08/27/2010] [Accepted: 09/23/2010] [Indexed: 02/07/2023]
Abstract
Apoptotic cell (AC)-derived factors alter the physiology of macrophages (MΦs) towards a regulatory phenotype, characterized by reduced nitric oxide (NO) production. Impaired NO formation in response to AC-conditioned medium (CM) was facilitated by arginase II (ARG II) expression, which competes with inducible NO synthase for L-arginine. Here we explored signaling pathways allowing CM to upregulate ARG II in RAW264.7 MΦs. Sphingosine-1-phosphate (S1P) was required and acted synergistically with a so far unidentified factor to elicit high ARG II expression. S1P activated S1P(2), since S1P(2) knockdown prevented ARG II upregulation. Furthermore, ERK5 knockdown attenuated CM-mediated ARG II protein induction. CREB was implicated as shown by EMSA analysis and decoy-oligonucleotides scavenging CREB in RAW264.7 MΦs, which blocked ARG II expression. We conclude that AC-derived S1P binds to S1P(2) and acts synergistically with other factors to activate ERK5 and concomitantly CREB. This signaling cascade shapes an anti-inflammatory MΦ phenotype by ARG II induction.
Collapse
Affiliation(s)
- Vera Barra
- Faculty of Medicine, Institute of Biochemistry I, Goethe University Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany
| | - Anne-Marie Kuhn
- Faculty of Medicine, Institute of Biochemistry I, Goethe University Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany
| | - Andreas von Knethen
- Faculty of Medicine, Institute of Biochemistry I, Goethe University Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany
| | - Andreas Weigert
- Faculty of Medicine, Institute of Biochemistry I, Goethe University Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany
| | - Bernhard Brüne
- Faculty of Medicine, Institute of Biochemistry I, Goethe University Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany
| |
Collapse
|
24
|
Salomone S, Soydan G, Ip PCT, Hopson KMP, Waeber C. Vessel-specific role of sphingosine kinase 1 in the vasoconstriction of isolated basilar arteries. Pharmacol Res 2010; 62:465-74. [PMID: 20850539 DOI: 10.1016/j.phrs.2010.09.002] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/05/2010] [Revised: 09/07/2010] [Accepted: 09/08/2010] [Indexed: 12/20/2022]
Abstract
Sphingosine-1-phosphate (S1P) constricts cerebral arteries through S1P(3) receptor stimulation. Because the activity of the key S1P-synthesizing enzyme, sphingosine kinase (SPK), can be stimulated by agonists of various G protein-coupled receptors, it is likely that S1P also acts as a second messenger for other vasoconstrictors. We investigated the effect of SPK inhibitors and SPK gene deletion on the contractile responses of isolated vessels to vasoactive agonists and KCl-induced depolarization. Basilar and femoral arteries of rat, mounted in a wire myograph, were incubated with dimethylsphingosine (DMS), 2-(p-hydroxyanilino)-4-(p-chlorophenyl) thiazole (Compound 2) or FTY720, and exposed to KCl, 5-hydroxytryptamine (5-HT), S1P or phenylephrine (PE). Vasomotor responses in basilar artery were decreased by DMS, Compound 2 and FTY720, while they were not affected in femoral artery. Basilar arteries from SPK1(-/-) mice exhibited weaker vasoconstriction to both KCl and agonists (S1P and the prostanoid U46619) when compared to either wild type (WT) or SPK2(-/-). In contrast, in mesenteric resistance arteries, neither the contraction to KCl nor the maximum contraction to PE and S1P significantly differed among WT, SPK1(-/-) and SPK2(-/-). Quantitative analysis of SPK mRNA (reverse transcription and real time polymerase chain reaction) in mouse arteries showed 40-80-fold higher SPK1 expression in cerebral arteries than in aorta or mesenteric arteries. SPK1 critically modulates the reactivity of cerebral vasculature to vasoconstrictors. S1P plays a specific role as modulator of cerebral blood flow, potentially acting either directly outside vascular smooth muscle cells on S1P(3) receptors, or indirectly after being generated inside the cell in response to vasoconstrictors.
Collapse
Affiliation(s)
- Salvatore Salomone
- Stroke and Neurovascular Laboratory Regulation, Department of Radiology, Massachusetts General Hospital, Charlestown, MA, USA
| | | | | | | | | |
Collapse
|
25
|
Blom T, Bergelin N, Meinander A, Löf C, Slotte JP, Eriksson JE, Törnquist K. An autocrine sphingosine-1-phosphate signaling loop enhances NF-kappaB-activation and survival. BMC Cell Biol 2010; 11:45. [PMID: 20573281 PMCID: PMC2906432 DOI: 10.1186/1471-2121-11-45] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2009] [Accepted: 06/24/2010] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND Sphingosine-1-phosphate (S1P) is a bioactive lipid that regulates a multitude of cellular functions, including cell proliferation, survival, migration and angiogenesis. S1P mediates its effects either by signaling through G protein-coupled receptors (GPCRs) or through an intracellular mode of action. In this study, we have investigated the mechanism behind S1P-induced survival signalling. RESULTS We found that S1P protected cells from FasL-induced cell death in an NF-kappaB dependent manner. NF-kappaB was activated by extracellular S1P via S1P2 receptors and Gi protein signaling. Our study also demonstrates that extracellular S1P stimulates cells to rapidly produce and secrete additional S1P, which can further amplify the NF-kappaB activation. CONCLUSIONS We propose a self-amplifying loop of autocrine S1P with capacity to enhance cell survival. The mechanism provides increased understanding of the multifaceted roles of S1P in regulating cell fate during normal development and carcinogenesis.
Collapse
Affiliation(s)
- Tomas Blom
- Department of Biology, Abo Akademi University, 20520 Turku, Finland
| | | | | | | | | | | | | |
Collapse
|
26
|
Snider AJ, Orr Gandy KA, Obeid LM. Sphingosine kinase: Role in regulation of bioactive sphingolipid mediators in inflammation. Biochimie 2010; 92:707-15. [PMID: 20156522 DOI: 10.1016/j.biochi.2010.02.008] [Citation(s) in RCA: 133] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2009] [Accepted: 02/09/2010] [Indexed: 12/15/2022]
Abstract
Sphingolipids and their synthetic enzymes are emerging as important mediators in inflammatory responses and as regulators of immune cell functions. In particular, sphingosine kinase (SK) and its product sphingosine-1-phosphate (S1P) have been extensively implicated in these processes. SK catalyzes the phosphorylation of sphingosine to S1P and exists as two isoforms, SK1 and SK2. SK1 has been shown to be activated by cytokines including tumor necrosis factor-alpha (TNF-alpha) and interleukin1-beta (IL1-beta). The activation of SK1 in this pathway has been shown to be, at least in part, required for mediating TNF-alpha and IL1-beta inflammatory responses in cells, including induction of cyclo-oxygenase 2 (COX2). In addition to their role in inflammatory signaling, SK and S1P have also been implicated in various immune cell functions including, mast cell degranulation, migration of neutrophils, and migration and maturation of lymphocytes. The involvement of sphingolipids and sphingolipid metabolizing enzymes in inflammatory signaling and immune cell functions has implicated these mediators in numerous inflammatory disease states as well. The contribution of these mediators, specifically SK1 and S1P, to inflammation and disease are discussed in this review.
Collapse
Affiliation(s)
- Ashley J Snider
- Department of Medicine, Medical University of South Carolina, Charleston, SC 29403, USA
| | | | | |
Collapse
|
27
|
Hengst JA, Guilford JM, Conroy EJ, Wang X, Yun JK. Enhancement of sphingosine kinase 1 catalytic activity by deletion of 21 amino acids from the COOH-terminus. Arch Biochem Biophys 2010; 494:23-31. [PMID: 19914200 PMCID: PMC2812673 DOI: 10.1016/j.abb.2009.11.006] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2009] [Revised: 11/06/2009] [Accepted: 11/08/2009] [Indexed: 12/13/2022]
Abstract
Sphingosine kinase 1 (SphK1) responds to a variety of growth factor signals by increasing catalytic activity as it translocates to the plasma membrane (PM). Several studies have identified amino acids residues involved in translocation yet how SphK1 increases its catalytic activity remains to be elucidated. Herein, we report that deletion of 21 amino acids from the COOH-terminus of SphK1 (1-363) results in increased catalytic activity relative to wild-type SphK1 (1-384) which is independent of the phosphorylation state of Serine 225 and PMA stimulation. Importantly, HEK293 cells stably expressing the 1-363 protein exhibit enhanced cell growth under serum-deprived cell culture conditions. Together the evidence indicates that the COOH-terminal region of SphK1 encompasses a structural element that is necessary for the increase in catalytic activity in response to PMA treatment and that its deletion renders SphK1 constitutively active with respect to PMA treatment.
Collapse
Affiliation(s)
- Jeremy A Hengst
- Department of Pharmacology, The Pennsylvania State University College of Medicine, 500 University Drive, Hershey, PA, USA
| | | | | | | | | |
Collapse
|
28
|
Wong L, Tan SSL, Lam Y, Melendez AJ. Synthesis and evaluation of sphingosine analogues as inhibitors of sphingosine kinases. J Med Chem 2009; 52:3618-26. [PMID: 19469544 DOI: 10.1021/jm900121d] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Sphingolipid-metabolizing enzymes control the critical balance of the cellular levels of sphingolipids, including the apoptotic inducing ceramide (Cer) and the proliferative inducing sphingosine 1-phosphate (S1P). The production of S1P, catalyzed by the action of sphingosine kinases (SPHKs), is known to be critical for many cellular processes. However, it is suggested that SPHK, and/or its catalytic product S1P, plays critical roles in various diseases including autoimmune diseases, cancer, and allergies. However, there is a great limitation of specific pharmacological inhibitors for SPHKs. In this paper, we describe a novel and stereoselective method of synthesizing SPHKs inhibitors. We generated a number of novel compounds and identified a number of specific inhibitors of human SPHKs. These compounds demonstrated inhibition of SPHKs at micromolar concentrations, making them more potent than dimethylsphingosine (DMS), a well-known inhibitor of SPHKs. In particular, one of the inhibitors was found to be selective toward a particular isoform of SPHK.
Collapse
Affiliation(s)
- Lingkai Wong
- Department of Chemistry, Faculty of Science, National University of Singapore, 3 Science Drive 3, 117543, Singapore
| | | | | | | |
Collapse
|
29
|
Gαq-mediated plasma membrane translocation of sphingosine kinase-1 and cross-activation of S1P receptors. Biochim Biophys Acta Mol Cell Biol Lipids 2009; 1791:357-70. [DOI: 10.1016/j.bbalip.2009.01.019] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
30
|
Yu H, Okada T, Kobayashi M, Abo-Elmatty DM, Jahangeer S, Nakamura SI. Roles of extracellular and intracellular sphingosine 1-phosphate in cell migration. Genes Cells 2009; 14:597-605. [PMID: 19371379 DOI: 10.1111/j.1365-2443.2009.01295.x] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Sphingosine 1-phosphate (S1P) is an important factor for the regulation of cell motility acting both inside and outside the cells. The precise role of S1P in the control of cell motility, however, remains unclear. Here we describe the roles of S1P in the regulation of cell motility by dissecting them into intracellular and extracellular actions using a liposomal S1P transfer technique. In a Boyden chamber assay free S1P enhanced directional cell movement, whereas liposomal S1P induced nondirectional cell movement. Furthermore, inhibition of sphingosine kinase (SphK) 1 by several inhibitors or knockdown of the enzyme expression by siRNA caused reduced wound-faced cell polarity formation as assessed by wound-healing assay. Moreover, S1P-induced cell migration was strongly inhibited by SphK inhibitors. These results indicate that extracellular S1P acting through S1P receptors facilitates the formation of cell polarity, whereas S1P generated inside the cells functions as an intracellular mediator per se to enhance nondirectional cell movement, thus S1P enhances directional cell movement in a coordinated fashion.
Collapse
Affiliation(s)
- Huan Yu
- Division of Biochemistry, Department of Molecular and Cellular Biology, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan
| | | | | | | | | | | |
Collapse
|
31
|
Billich A, Urtz N, Reuschel R, Baumruker T. Sphingosine kinase 1 is essential for proteinase-activated receptor-1 signalling in epithelial and endothelial cells. Int J Biochem Cell Biol 2009; 41:1547-55. [PMID: 19162217 DOI: 10.1016/j.biocel.2009.01.001] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2008] [Revised: 12/24/2008] [Accepted: 01/05/2009] [Indexed: 01/07/2023]
Abstract
There is accumulating evidence that activation of sphingosine kinase 1 (SPHK1) is an important element in intracellular signalling cascades initiated by stimulation of multiple receptors, including certain growth factor, cytokine, and also G-protein coupled receptors. We here report that stimulation of the lung epithelial cell line A549 by thrombin leads to transient increase of SPHK1 activity and elevation of intracellular sphingosine-1-phosphate (S1P); abrogation of this stimulation by SPHK1-specific siRNA, pharmacological inhibition, or expression of a dominant-negative SPHK1 mutant blocks the response to thrombin, as measured by secretion of MCP-1, IL-6, IL-8, and PGE(2). Using selective stimulation of proteinase-activated receptors (PARs) a specific involvement of SPHK1 in the PAR-1 induced responses in A549 cell, including activation of NFkappaB, was evident, while PAR-2 and PAR-4 responses were independent of SPHK1. Moreover, PAR-1 or thrombin-induced cytokine production and adhesion factor expression of human umbilical vein endothelial cells was also seen to depend on SPHK1. Using dermal microvascular endothelial cells from SPHK1-deficient mice, we showed that absence of the enzyme abrogates MCP-1 production induced in these cells upon treatment with thrombin or PAR-1 activating peptide. We propose SPHK1 inhibition as a novel way to block PAR-1 mediated signalling, which could be useful in treatment of a number of diseases, in particular in atherosclerosis.
Collapse
Affiliation(s)
- Andreas Billich
- Novartis Institutes for BioMedical Research, Brunnerstrasse 59, Vienna, Austria.
| | | | | | | |
Collapse
|
32
|
K6PC-5, a Direct Activator of Sphingosine Kinase 1, Promotes Epidermal Differentiation Through Intracellular Ca2+ Signaling. J Invest Dermatol 2008; 128:2166-78. [DOI: 10.1038/jid.2008.66] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
33
|
Lichte K, Rossi R, Danneberg K, Braak MT, Kürschner U, Jakobs KH, Kleuser B, Heringdorf DMZ. Lysophospholipid Receptor-Mediated Calcium Signaling in Human Keratinocytes. J Invest Dermatol 2008; 128:1487-98. [DOI: 10.1038/sj.jid.5701207] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
34
|
Takabe K, Paugh SW, Milstien S, Spiegel S. "Inside-out" signaling of sphingosine-1-phosphate: therapeutic targets. Pharmacol Rev 2008; 60:181-95. [PMID: 18552276 PMCID: PMC2695666 DOI: 10.1124/pr.107.07113] [Citation(s) in RCA: 569] [Impact Index Per Article: 33.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Sphingosine 1-phosphate (S1P) is a bioactive sphingolipid metabolite involved in many critical cellular processes including proliferation, survival, and migration, as well as angiogenesis and allergic responses. S1P levels inside cells are tightly regulated by the balance between its synthesis by sphingosine kinases and degradation. S1P is interconvertible with ceramide, which is a critical mediator of apoptosis. It has been postulated that the ratio between S1P and ceramide determines cell fate. Activation of sphingosine kinase by a variety of agonists increases intracellular S1P, which in turn can function intracellularly as a second messenger or be secreted out of the cell and act extracellularly by binding to and signaling through S1P receptors in autocrine and/or paracrine manners. Recent studies suggest that this "inside-out" signaling by S1P may play a role in many human diseases, including cancer, atherosclerosis, inflammation, and autoimmune disorders such as multiple sclerosis. In this review we summarize metabolism of S1P, mechanisms of sphingosine kinase activation, and S1P receptors and their downstream signaling pathways and examine relationships to multiple disease processes. In particular, we describe recent preclinical and clinical trials of therapies targeting S1P signaling, including 2-amino-2-propane-1,3-diol hydrochloride (FTY720, fingolimod), S1P receptor agonists, sphingosine kinase inhibitors, and anti-S1P monoclonal antibody.
Collapse
Affiliation(s)
- Kazuaki Takabe
- Department of Surgery, Division of Surgical Oncology, Virginia Commonwealth University School of Medicine, Richmond, VA 23298, USA
| | | | | | | |
Collapse
|
35
|
K6PC-5, a sphingosine kinase activator, induces anti-aging effects in intrinsically aged skin through intracellular Ca2+ signaling. J Dermatol Sci 2008; 51:89-102. [PMID: 18420384 DOI: 10.1016/j.jdermsci.2008.03.002] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2007] [Revised: 02/20/2008] [Accepted: 03/03/2008] [Indexed: 01/01/2023]
Abstract
BACKGROUND Sphingosine-1-phosphate (S1P), a bioactive sphingolipid metabolite, regulates multiple cellular responses such as Ca(2+) signaling, growth, survival, and differentiation. Because sphingosine kinase (SK) is the enzyme directly responsible for the production of S1P, many factors have been identified that regulate its activity and subsequent S1P levels. To date, there are no reports to demonstrate a chemically induced, direct activation of SK. OBJECTIVE Here we have studied the effects of K6PC-5 as a newly synthesized SK activator on fibroblast proliferation in both human fibroblasts and aged mouse skin. To demonstrate that K6PC-5 has S1P-mediated action mechanism in fibroblasts, we have measured SK-dependent intracellular Ca(2+) signaling. METHODS Fibroblasts were cultured primarily from human foreskin and were used to study the effect of K6PC-5 and S1P on intracellular Ca(2+) signaling and fibroblast proliferation. Changes in intracellular Ca(2+) were detected by fluorescence with fura-2/AM. To study skin anti-aging effects of K6PC-5, we used intrinsically aged hairless mice (56 weeks old). RESULTS K6PC-5 promoted fibroblast proliferation and procollagen production in human fibroblasts significantly. K6PC-5 induced intracellular Ca(2+) concentration ([Ca(2+)](i)) oscillations in human fibroblasts. Both dimethylsphingosine and dihydroxysphingosine, SK inhibitors, and the transfection of SK1-siRNA blocked the K6PC-5-induced increases in [Ca(2+)](i), an effect independent of the classical PLC/IP(3)-mediated pathway. The K6PC-5-induced [Ca(2+)](i) oscillations were dependent on thapsigargin-sensitive Ca(2+) stores and Ca(2+) entry. Topical application of K6PC-5 for 2 weeks to intrinsically aged hairless mice enhanced fibroblast proliferation, collagen production, and eventually increased dermal thickness (10%). K6PC-5 also promoted specific epidermal differentiation marker proteins, including involucrin, loricrin, filaggrin, and keratin 5, without any alterations on epidermal barrier function. CONCLUSION These results suggest that K6PC-5 acts to regulate fibroblast proliferation through intracellular S1P production, and can further promote keratinocyte differentiation. We anticipate that the regulation of S1P levels may represent a novel approach for the treatment of skin disorders, including skin aging.
Collapse
|
36
|
Eskan MA, Rose BG, Benakanakere MR, Lee MJ, Kinane DF. Sphingosine 1-phosphate 1 and TLR4 mediate IFN-beta expression in human gingival epithelial cells. THE JOURNAL OF IMMUNOLOGY 2008; 180:1818-25. [PMID: 18209079 DOI: 10.4049/jimmunol.180.3.1818] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
IFN-beta production is a critical step in human innate immune responses and is primarily controlled at the transcription level by highly ordered mechanisms. IFN-beta can be induced by pattern-recognition receptors such as the TLR4. S1P1 is a G protein-coupled receptor, which has a high affinity for sphingosine 1-phosphate (S1P). Although many of the receptors and signaling pathways leading to the expression of IFN-beta have been identified and characterized, it is still unclear how IFN-beta is regulated in primary human gingival epithelial cells (HGECs). In this study, we demonstrate that S1P1 and TLR4, acting in unison, play an important role in IFN-beta expression at the protein and mRNA level in HGECs. We demonstrate that the expression of both IFN-beta and IFN-inducible protein-10 (CXCL-10) is significantly up-regulated by LPS and S1P or LPS and a specific S1P1 agonist. This enhanced innate immune response is attenuated in HGECs by small interfering RNA knockdown of either TLR4 or S1P1. Moreover, we show that triggering of TLR4 results in the increased expression of S1P1 receptors. Furthermore, we found that IFN-regulatory factor 3 activation was maximized by LPS and S1P through PI3K. Our data show that triggering TLR4 increases S1P1, such that both TLR4 and S1P1 acting through PI3K enhancement of IFN-regulatory factor 3 activation increase IFN-beta expression in epithelial cells. The functional association between TLR4 and the S1P1 receptor demonstrates a novel mechanism in the regulation of IFN-beta and CXCL-10 in human primary gingival epithelial cells.
Collapse
Affiliation(s)
- Mehmet A Eskan
- Center for Oral Health and Systemic Disease, Department of Periodontics, Endodontics, and Dental Hygiene, University of Louisville School of Dentistry, 501 South Preston Street, Louisville, KY 40202, USA
| | | | | | | | | |
Collapse
|
37
|
Alvarez SE, Milstien S, Spiegel S. Autocrine and paracrine roles of sphingosine-1-phosphate. Trends Endocrinol Metab 2007; 18:300-7. [PMID: 17904858 DOI: 10.1016/j.tem.2007.07.005] [Citation(s) in RCA: 246] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/16/2007] [Revised: 07/18/2007] [Accepted: 07/18/2007] [Indexed: 11/16/2022]
Abstract
Sphingosine-1-phosphate (S1P) is a bioactive sphingolipid metabolite that has been implicated in many biological processes, including cell migration, survival, proliferation, angiogenesis and immune and allergic responses. S1P levels inside cells are regulated tightly by the balance between its synthesis by sphingosine kinases and degradation by S1P lyases and S1P phosphatases. Activation of sphingosine kinase by any of a variety of agonists increases S1P levels, which in turn can function intracellularly as a second messenger or in an autocrine and/or paracrine fashion to activate and signal through S1P receptors present on the surface of the cell. This review summarizes recent findings on the roles of S1P as a mediator of the actions of cytokines, growth factors and hormones.
Collapse
Affiliation(s)
- Sergio E Alvarez
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University School of Medicine, Richmond, VA 23298, USA
| | | | | |
Collapse
|
38
|
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
|
39
|
Roviezzo F, Di Lorenzo A, Bucci M, Brancaleone V, Vellecco V, De Nardo M, Orlotti D, De Palma R, Rossi F, D'Agostino B, Cirino G. Sphingosine-1-phosphate/sphingosine kinase pathway is involved in mouse airway hyperresponsiveness. Am J Respir Cell Mol Biol 2007; 36:757-62. [PMID: 17322125 DOI: 10.1165/rcmb.2006-0383oc] [Citation(s) in RCA: 88] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Sphingosine-1-phosphate (S1P) has been shown to regulate numerous and diverse cell functions, including smooth muscle contraction. Here we assessed the role of S1P/Sphingosine kinase (SPK) pathway in the regulation of bronchial tone. Our objective was to determine, using an integrated pharmacologic and molecular approach, (1) the role of S1P as endogenous modulator of the bronchial tone, and (2) the linkage between S1P pathway and bronchial hyperresponsiveness. We evaluated S1P effects on isolated bronchi and whole lungs, harvested from Balb/c mice sensitized to ovalbumin (OVA) versus vehicle-treated mice, by measuring bronchial reactivity and lung resistance. We found that S1P administration on nonsensitized mouse bronchi does not cause any direct effect on bronchial tone, while a significant increase in Ach-induced contraction occurs after S1P challenge. Conversely, in OVA-sensitized mice S1P/SPK pathway triggers airway hyperesponsiveness. Indeed, S1P causes a dose-dependent contraction of isolated bronchi. Similarly, in the whole lung system S1P increased airway resistance only in OVA-sensitized mice. The action on bronchi of S1P is coupled to an enhanced expression of SPK(1) and SPK(2) as well as of S1P(2) and S1P(3) receptors. In these experiments the key role for S1P/SPK in hyperreactivity has been confirmed by pharmacologic modulation of SPKs. S1P/SPK pathway does not seem to play a major role in physiologic conditions, while it may become critical in pathologic conditions. These results open new windows for therapeutic strategies in diseases like asthma.
Collapse
Affiliation(s)
- Fiorentina Roviezzo
- Dipartimento di Farmacologia Sperimentale, Università di Napoli Federico II, Napoli, Italy
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Abstract
Sphingosine kinase-1 (SphK1) catalyses the phosphorylation of sphingosine to sphingosine-1-phosphate (S1P), which acts on at least five specific G-protein-coupled receptors and also intracellularly. SphK1 has been implicated in cell proliferation, cancer growth, chemoresistance, immune cell functions and cell migration. In this issue of the British Journal of Pharmacology, Klawitter et al. demonstrate that extracellular nucleotides stimulate the migration of renal mesangial cells. The nucleotides furthermore upregulated SphK1 expression and activity, and this enzyme was required for nucleotide-induced migration. Together with previous findings, these data raise exciting questions: by which mechanism does SphK1 regulate migration in mesangial cells, how is the interplay of purinoceptors and S1P receptors organized in these cells, and how would SphK1-deficient mice respond to kidney damage?
Collapse
|
41
|
El-Shewy HM, Johnson KR, Lee MH, Jaffa AA, Obeid LM, Luttrell LM. Insulin-like Growth Factors Mediate Heterotrimeric G Protein-dependent ERK1/2 Activation by Transactivating Sphingosine 1-Phosphate Receptors. J Biol Chem 2006. [DOI: 10.1016/s0021-9258(19)84052-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
|
42
|
El-Shewy HM, Johnson KR, Lee MH, Jaffa AA, Obeid LM, Luttrell LM. Insulin-like growth factors mediate heterotrimeric G protein-dependent ERK1/2 activation by transactivating sphingosine 1-phosphate receptors. J Biol Chem 2006; 281:31399-407. [PMID: 16926156 DOI: 10.1074/jbc.m605339200] [Citation(s) in RCA: 68] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Although several studies have shown that a subset of insulin-like growth factor (IGF) signals require the activation of heterotrimeric G proteins, the molecular mechanisms underlying IGF-stimulated G protein signaling remain poorly understood. Here, we have studied the mechanism by which endogenous IGF receptors activate the ERK1/2 mitogen-activated protein kinase cascade in HEK293 cells. In these cells, treatment with pertussis toxin and expression of a Galpha(q/11)-(305-359) peptide that inhibits G(q/11) signaling additively inhibited IGF-stimulated ERK1/2 activation, indicating that the signal was almost completely G protein-dependent. Treatment with IGF-1 or IGF-2 promoted translocation of green fluorescent protein (GFP)-tagged sphingosine kinase (SK) 1 from the cytosol to the plasma membrane, increased endogenous SK activity within 30 s of stimulation, and caused a statistically significant increase in intracellular and extracellular sphingosine 1-phosphate (S1P) concentration. Using a GFP-tagged S1P1 receptor as a biological sensor for the generation of physiologically relevant S1P levels, we found that IGF-1 and IGF-2 induced GFP-S1P receptor internalization and that the effect was blocked by pretreatment with the SK inhibitor, dimethylsphingosine. Treating cells with dimethylsphingosine, silencing SK1 expression by RNA interference, and blocking endogenous S1P receptors with the competitive antagonist VPC23019 all significantly inhibited IGF-stimulated ERK1/2 activation, suggesting that IGFs elicit G protein-dependent ERK1/2 activation by stimulating SK1-dependent transactivation of S1P receptors. Given the ubiquity of SK and S1P receptor expression, S1P receptor transactivation may represent a general mechanism for G protein-dependent signaling by non-G protein-coupled receptors.
Collapse
Affiliation(s)
- Hesham M El-Shewy
- Department of Medicine, Medical University of South Carolina, Charleston, South Carolina 29425, USA
| | | | | | | | | | | |
Collapse
|
43
|
Paugh SW, Cassidy MP, He H, Milstien S, Sim-Selley LJ, Spiegel S, Selley DE. Sphingosine and its analog, the immunosuppressant 2-amino-2-(2-[4-octylphenyl]ethyl)-1,3-propanediol, interact with the CB1 cannabinoid receptor. Mol Pharmacol 2006; 70:41-50. [PMID: 16571654 DOI: 10.1124/mol.105.020552] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Sphingosine-1-phosphate (S1P) and cannabinoid receptors are G-protein-coupled receptors that mediate the effects of S1P and endocannabinoids, respectively. Cannabinoid receptors also mediate the effects of Delta9-tetrahydrocannabinol, the primary psychoactive ingredient in marijuana, whereas S1P receptors contribute to the immunosuppressant effects of 2-amino-2-(2-[4-octylphenyl]ethyl)-1,3-propanediol (FTY720). FTY720 is a sphingosine analog that can prevent renal graft rejections and suppress a variety of autoimmune disorders in animal models and clinical trials. We now report that both FTY720 and sphingosine interact with CB1 but not CB2 cannabinoid receptors. FTY720 and sphingosine inhibited the binding of the CB1-selective antagonist [3H]N-(piperidinyl)-5-(4-chlorophenyl)-1-(2,4-dichlorophenyl)-4-methyl-1H-pyrazole-3-carboxamide ([3H]SR141716A) and the cannabinoid agonist [3H](-)-cis-3-[2-hydroxy-4-(1,1-dimethylheptyl)phenyl]-trans-4-(3-hydroxypropyl)cyclohexanol ([3H]CP55,940) in a concentration-dependent manner in both CB1-expressing cell lines and mouse cerebellum. However, these compounds did not significantly alter [3H]CP55,940 binding to CB2 receptors. In G-protein activation assays, FTY720 and sphingosine inhibited the maximal stimulation of guanosine 5'-O-(3-[35S]thio)triphosphate binding by the cannabinoid agonist R-(+)-[2,3-dihydro-5-methyl-3-[(morpholinyl)methyl]pyrrolo[1,2,3-de]-1,4-benzoxazinyl]-(1-naphthalenyl)methanone mesylate (WIN55,212-2) in a concentration-dependent manner, and this antagonist effect was not mimicked by S1P. FTY720 and sphingosine also inhibited activation of extracellular signal-regulated kinases 1 and 2 and Akt by WIN55,212-2 in intact Chinese hamster ovary (CHO) cells expressing CB1 receptors and attenuated WIN55,212-2-stimulated internalization of a fluorescence-tagged CB1 receptor in CHO cells. Moreover, both FTY720 and sphingosine produced rightward shifts in the concentration-effect curves of cannabinoid agonists for G-protein activation, indicating that they act as competitive CB1 antagonists. These results suggest that the CB1 receptor could be a novel target of FTY720 and that sphingosine could be an endogenous CB11 antagonist.
Collapse
MESH Headings
- Animals
- Benzoxazines
- Binding, Competitive/drug effects
- CHO Cells
- Cell Line
- Cricetinae
- Cricetulus
- Cyclohexanes/metabolism
- Cyclohexanes/pharmacology
- Cyclohexanols
- Endocytosis/drug effects
- Enzyme Activation/drug effects
- Fingolimod Hydrochloride
- Guanosine 5'-O-(3-Thiotriphosphate)/metabolism
- Humans
- Immunosuppressive Agents/metabolism
- Immunosuppressive Agents/pharmacology
- Mice
- Mitogen-Activated Protein Kinase 1/metabolism
- Mitogen-Activated Protein Kinase 3/metabolism
- Morpholines/metabolism
- Morpholines/pharmacology
- Naphthalenes/metabolism
- Naphthalenes/pharmacology
- Phenols/metabolism
- Phenols/pharmacology
- Piperidines/metabolism
- Piperidines/pharmacology
- Propylene Glycols/metabolism
- Propylene Glycols/pharmacology
- Proto-Oncogene Proteins c-akt/metabolism
- Pyrazoles/metabolism
- Pyrazoles/pharmacology
- Radioligand Assay
- Receptor, Cannabinoid, CB1/agonists
- Receptor, Cannabinoid, CB1/antagonists & inhibitors
- Receptor, Cannabinoid, CB1/metabolism
- Receptor, Cannabinoid, CB2/agonists
- Receptor, Cannabinoid, CB2/antagonists & inhibitors
- Receptor, Cannabinoid, CB2/metabolism
- Rimonabant
- Sphingosine/analogs & derivatives
- Sphingosine/metabolism
- Sphingosine/pharmacology
Collapse
Affiliation(s)
- Steven W Paugh
- Department of Pharmacology and Toxicology, Virginia Commonwealth University School of Medicine, 1112 East Clay Street, Box 980524, Richmond, VA 23298, USA
| | | | | | | | | | | | | |
Collapse
|
44
|
Wattenberg BW, Pitson SM, Raben DM. The sphingosine and diacylglycerol kinase superfamily of signaling kinases: localization as a key to signaling function. J Lipid Res 2006; 47:1128-39. [PMID: 16520486 DOI: 10.1194/jlr.r600003-jlr200] [Citation(s) in RCA: 104] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
The sphingosine and diacylglycerol kinases form a superfamily of structurally related lipid signaling kinases. One of the striking features of these kinases is that although they are clearly involved in agonist-mediated signaling, this signaling is accomplished with only a moderate (and sometimes no) increase in the enzymatic activity of the enzymes. Here, we summarize findings that indicate that signaling by these kinases is strongly dependent on their localization to specific intracellular sites rather than on increases in enzyme activity. Both the substrates and products of these enzymes are bioactive lipids. Moreover, many of the metabolic enzymes that act on these lipids are found in specific organelles. Therefore, changes in the membrane localization of these signaling kinases have profound effects not only on the production of signaling lipid phosphates but also on the metabolism of the upstream signaling lipids.
Collapse
|
45
|
Xu SZ, Muraki K, Zeng F, Li J, Sukumar P, Shah S, Dedman AM, Flemming PK, McHugh D, Naylor J, Cheong A, Bateson AN, Munsch CM, Porter KE, Beech DJ. A sphingosine-1-phosphate-activated calcium channel controlling vascular smooth muscle cell motility. Circ Res 2006; 98:1381-9. [PMID: 16675717 PMCID: PMC2648505 DOI: 10.1161/01.res.0000225284.36490.a2] [Citation(s) in RCA: 125] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
In a screen of potential lipid regulators of transient receptor potential (TRP) channels, we identified sphingosine-1-phosphate (S1P) as an activator of TRPC5. We explored the relevance to vascular biology because S1P is a key cardiovascular signaling molecule. TRPC5 is expressed in smooth muscle cells of human vein along with TRPC1, which forms a complex with TRPC5. Importantly, S1P also activates the TRPC5-TRPC1 heteromultimeric channel. Because TRPC channels are linked to neuronal growth cone extension, we considered a related concept for smooth muscle. We find S1P stimulates smooth muscle cell motility, and that this is inhibited by E3-targeted anti-TRPC5 antibody. Ion permeation involving TRPC5 is crucial because S1P-evoked motility is also suppressed by the channel blocker 2-aminoethoxydiphenyl borate or a TRPC5 ion-pore mutant. S1P acts on TRPC5 via two mechanisms, one extracellular and one intracellular, consistent with its bipolar signaling functions. The extracellular effect appears to have a primary role in S1P-evoked cell motility. The data suggest S1P sensing by TRPC5 calcium channel is a mechanism contributing to vascular smooth muscle adaptation.
Collapse
Affiliation(s)
- Shang-Zhong Xu
- Institute of Membrane and Systems Biology, University of Leeds, Leeds, LS2 9JT, UK
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Heacock AM, Dodd MS, Fisher SK. Regulation of volume-sensitive osmolyte efflux from human SH-SY5Y neuroblastoma cells following activation of lysophospholipid receptors. J Pharmacol Exp Ther 2006; 317:685-93. [PMID: 16415087 DOI: 10.1124/jpet.105.098467] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The ability of the lysophospholipids sphingosine 1-phosphate (S1P) and lysophosphatidic acid (LPA) to promote the release of the organic osmolyte taurine in response to hypoosmotic stress has been examined. Incubation of SH-SY5Y neuroblastoma cells under hypoosmotic conditions (230 mOsM) resulted in a time-dependent release of taurine that was markedly enhanced (3-7-fold) by the addition of micromolar concentrations of either S1P or LPA. At optimal concentrations, the effects of S1P and LPA on taurine efflux were additive and mediated via distinct receptors. Inclusion of 1,9-dideoxyfoskolin, 5-nitro-2-(3-phenylpropylamino benzoic acid, or 4-[(2-butyl-6,7-dichloro-2-cyclopentyl-2,3-dihydro-1-oxo-1H-inden-5-yl)oxy]-butanoic acid blocked the ability of both lysophospholipids to enhance taurine release, indicating the mediation of a volume-sensitive organic osmolyte and anion channel. Both S1P and LPA elicited robust increases in intracellular calcium concentration that were attenuated by the removal of extracellular calcium, abolished by the depletion of intracellular calcium with thapsigargin, and were independent of phosphoinositide turnover. Taurine efflux mediated by S1P and LPA was unaffected by the removal of extracellular calcium but was attenuated by depletion of intracellular calcium (34-38%) and by inhibition of protein kinase C (PKC) with chelerythrine (38-72%). When intracellular calcium was depleted and PKC was inhibited, S1P- or LPA-stimulated taurine efflux was inhibited by 80%. Pretreatment of the cells with pertussis toxin, toxin B, or cytochalasin D had no effect on lysophospholipid-stimulated taurine efflux. The results indicate that both S1P and LPA receptors facilitate osmolyte release via a phospholipase C-independent mechanism that requires the availability of intracellular calcium and PKC activity.
Collapse
Affiliation(s)
- Anne M Heacock
- University of Michigan, Molecular and Behavioral Neuroscience Institute Laboratories at MSRB II, 1150 West Medical Center Drive, C560, MSRB II, Ann Arbor, MI 48109-0669, USA
| | | | | |
Collapse
|
47
|
Hemmings DG. Signal transduction underlying the vascular effects of sphingosine 1-phosphate and sphingosylphosphorylcholine. Naunyn Schmiedebergs Arch Pharmacol 2006; 373:18-29. [PMID: 16570136 DOI: 10.1007/s00210-006-0046-5] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Two related lysosphingolipids, sphingosine 1-phosphate (S1P) and sphingosylphosphorylcholine (SPC) mediate diverse cellular responses through signals transduced by either activation of G-protein coupled receptors or possibly by acting intracellularly. Vascular responses to S1P and SPC measured both in vivo and in dissected vessels show predominantly vasoconstriction with some evidence for vasodilation. Although stimulation with S1P or SPC generally leads to similar vascular responses, the signalling pathways stimulated to produce these responses are often distinct. Nevertheless, mobilization of Ca2+ from intracellular stores and influx of extracellular Ca2+, which both increase [Ca2+]i, occur in response to S1P and SPC. Both mobilization of Ca2+ from intracellular stores and influx of extracellular Ca2+ occur in response to S1P and SPC. As well, both S1P and SPC induce Ca2+-sensitization in vascular smooth muscle which is mediated through Rho kinase activation. In the endothelium, S1P and SPC stimulate the production of the vasodilator, nitric oxide through activation of endothelial nitric oxide synthase. This activation occurs through phosphorylation by Akt and through binding of Ca2+-calmodulin upon increased [Ca2+]i. These lysosphingolipids also activate cyclooxygenase-2 which produces prostaglandins with both vasoconstrictor and vasodilator properties. A balance between the signals inducing vasodilation versus the signals inducing vasoconstriction will determine the vascular outcome. Thus, perturbations in S1P and SPC concentrations, relative expression of receptors or downstream signalling pathways may provide a mechanism for pathophysiological conditions such as hypertension. Given this background, recent studies examining a potential role for S1P and SPC in hypertension and vascular dysfunction in aging are discussed.
Collapse
Affiliation(s)
- Denise G Hemmings
- Department Obstetrics and Gynecology, Perinatal Research Centre, University of Alberta, 227 Heritage Medical Research Center, T6G 2S2, Edmonton, Alberta, Canada.
| |
Collapse
|
48
|
Wendler CC, Rivkees SA. Sphingosine-1-phosphate inhibits cell migration and endothelial to mesenchymal cell transformation during cardiac development. Dev Biol 2006; 291:264-77. [PMID: 16434032 DOI: 10.1016/j.ydbio.2005.12.013] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2005] [Revised: 11/19/2005] [Accepted: 12/06/2005] [Indexed: 11/23/2022]
Abstract
Sphingosine-1-phosphate (S1P) is a biologically active sphingolipid metabolite that exerts important effects on numerous cellular events via cell surface receptors, S1P(1-5). S1P influences differentiation, proliferation, and migration during vascular development. However, the effects of S1P signaling on early cardiac development are not well understood. To address this issue, we examined the expression of S1P regulatory enzymes and S1P receptors during cardiac development. We observed that enzymes that regulate S1P levels, sphingosine kinase and sphingosine-1-phosphate phosphatase, are expressed in the developing heart. In addition, RT-PCR revealed that four of the five known S1P receptors (S1P(1-4)) are also expressed in the developing heart. Next, effects of altered S1P levels on whole embryo and atrioventricular (AV) canal cultures were investigated. We demonstrate that inactivation of the S1P producing enzyme, sphingosine kinase, leads to cell death in cardiac tissue which is rescued by exogenous S1P treatment. Other experiments reveal that increased S1P concentration prevents alterations in cell morphology that are required for cell migration. This effect results in reduced cell migration and inhibited mesenchymal cell formation in AV canal cushion tissue. These data indicate that S1P, locally maintained within a specific concentration range, is an important and necessary component of early heart development.
Collapse
Affiliation(s)
- Christopher C Wendler
- Section of Developmental Endocrinology and Biology, Yale Child Health Research Center, Department of Pediatrics, Yale University School of Medicine, New Haven, CT 06520, USA.
| | | |
Collapse
|
49
|
Inagaki Y, Pham T, Fujiwara Y, Kohno T, Osborne D, Igarashi Y, Tigyi G, Parrill A. Sphingosine 1-phosphate analogue recognition and selectivity at S1P4 within the endothelial differentiation gene family of receptors. Biochem J 2005; 389:187-95. [PMID: 15733055 PMCID: PMC1184551 DOI: 10.1042/bj20050046] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
Synergistic computational and experimental studies provided previously unforeseen details concerning the structural basis of S1P (sphingosine 1-phosphate) recognition by the S1P4 G-protein-coupled receptor. Similarly to reports on the S1P1 receptor, cationic and anionic residues in the third transmembrane domain (R3.28 and E3.29 at positions 124 and 125) form ion pairs with the phosphate and ammonium of S1P, and alanine mutations at these positions abolished specific S1P binding, S1P-induced receptor activation and cell migration. Unlike findings on the S1P1 receptor, no cationic residue in the seventh transmembrane domain interacts with the phosphate. Additionally, two previously undiscovered interactions with the S1P polar headgroup have been identified. Trp186 at position 4.64 in the fourth transmembrane domain interacts by a cation-pi interaction with the ammonium group of S1P. Lys204 at position 5.38 forms an ion pair with the S1P. The S1P4 and S1P1 receptors show differences in binding-pocket shape and electrostatic distributions that correlate with the published structure-activity relationships. In particular, the binding pocket of mS1P4 (mouse S1P4) has recognition sites for the anionic phosphate and cationic ammonium groups that are equidistant from the end of the non-polar tail. In contrast, the binding pocket of hS1P1 (human S1P4) places the ammonium recognition site 2 A (1 A=0.1 nm) closer to the end of the non-polar tail than the phosphate recognition site.
Collapse
Affiliation(s)
- Yuichi Inagaki
- *Department of Biomembrane and Biofunctional Chemistry, Graduate School of Pharmaceutical Sciences, Hokkaido University, Kita 12-jo, Nishi 6-choume, Kita-ku, Sapporo 060-0812, Japan
| | - TrucChi T. Pham
- †Department of Chemistry and Computational Research on Materials Institute, The University of Memphis, Memphis, TN 38152, U.S.A
| | - Yuko Fujiwara
- ‡Department of Physiology, University of Tennessee Health Sciences Center, Memphis, TN 38163, U.S.A
| | - Takayuki Kohno
- *Department of Biomembrane and Biofunctional Chemistry, Graduate School of Pharmaceutical Sciences, Hokkaido University, Kita 12-jo, Nishi 6-choume, Kita-ku, Sapporo 060-0812, Japan
| | - Daniel A. Osborne
- †Department of Chemistry and Computational Research on Materials Institute, The University of Memphis, Memphis, TN 38152, U.S.A
| | - Yasuyuki Igarashi
- *Department of Biomembrane and Biofunctional Chemistry, Graduate School of Pharmaceutical Sciences, Hokkaido University, Kita 12-jo, Nishi 6-choume, Kita-ku, Sapporo 060-0812, Japan
| | - Gabor Tigyi
- †Department of Chemistry and Computational Research on Materials Institute, The University of Memphis, Memphis, TN 38152, U.S.A
- ‡Department of Physiology, University of Tennessee Health Sciences Center, Memphis, TN 38163, U.S.A
| | - Abby L. Parrill
- †Department of Chemistry and Computational Research on Materials Institute, The University of Memphis, Memphis, TN 38152, U.S.A
- ‡Department of Physiology, University of Tennessee Health Sciences Center, Memphis, TN 38163, U.S.A
- To whom correspondence should be addressed (email )
| |
Collapse
|
50
|
Baumruker T, Bornancin F, Billich A. The role of sphingosine and ceramide kinases in inflammatory responses. Immunol Lett 2005; 96:175-85. [PMID: 15585321 DOI: 10.1016/j.imlet.2004.09.001] [Citation(s) in RCA: 58] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2004] [Accepted: 09/03/2004] [Indexed: 12/20/2022]
Abstract
The 1-phosphates of sphingosine and ceramide (S1P and C1P) have emerged as key representatives of a new group of lipid signalling molecules. S1P is known to act both as an extracellular mediator and as an intracellular 'second messenger,' while C1P currently is only known for its intracellular actions. Therefore, sphingosine and ceramide kinases, the enzymes involved in the generation of these lipid mediators, are now in the spotlight. This review summarizes current information on structure, localization, substrate specificity, activation, and binding partners of these kinases, and then focuses on discoveries in relation to immune cell regulation and inflammation, addressing in particular mast cell activation and degranulation, IL-12 signalling, prostaglandin biosynthesis, monocyte activation, and neutrophil priming.
Collapse
Affiliation(s)
- Thomas Baumruker
- Novartis Institute for BioMedical Research Vienna, Brunner Strasse 59, A-1235 Vienna, Austria.
| | | | | |
Collapse
|