1
|
Zhu W, Hu J, Li Y, Yang B, Guan Y, Xu C, Chen F, Chi J, Bao Y. Comparative Proteomic Analysis of Pleurotus ostreatus Reveals Great Metabolic Differences in the Cap and Stipe Development and the Potential Role of Ca 2+ in the Primordium Differentiation. Int J Mol Sci 2019; 20:ijms20246317. [PMID: 31847351 PMCID: PMC6940972 DOI: 10.3390/ijms20246317] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Revised: 12/06/2019] [Accepted: 12/11/2019] [Indexed: 01/08/2023] Open
Abstract
Pleurotus ostreatus is a widely cultivated edible fungus around the world. At present, studies on the developmental process of the fruiting body are limited. In our study, we compared the differentially expressed proteins (DEPs) in the stipe and cap of the fruiting body by high-throughput proteomics. GO and pathway analysis revealed the great differences in the metabolic levels, including sucrose and starch metabolism, and sphingolipid signaling and metabolism, and the differences of 16 important DEPs were validated further by qPCR analysis in expression level. In order to control the cap and stipe development, several chemical inducers were applied to the primordium of the fruiting body according to the pathway enrichment results. We found that CaCl2 can affect the primordium differentiation through inhibiting the stipe development. EGTA (ethyleneglycol bis (β-aminoethyl ether)-N,N,N′,N′-tetraacetic acid) treatment confirmed the inhibitory role of Ca2+ in the stipe development. Our study not only shows great metabolic differences during the cap and stipe development but also reveals the underlying mechanism directing the primordium differentiation in the early development of the fruiting body for the first time. Most importantly, we provide a reliable application strategy for the cultivation and improvement of the Pleurotus ostreatus, which can be an example and reference for a more edible fungus.
Collapse
Affiliation(s)
- Weiwei Zhu
- School of Bioengineering, Dalian University of Technology, Dalian 116024, China;
- Microbial Research Institute of Liaoning Province, Chaoyang 122000, China; (Y.L.); (Y.G.); (C.X.); (F.C.); (J.C.)
| | - Jinbo Hu
- Laboratory of Photosynthesis and Environment, CAS Center for Excellence in Molecular Plant Sciences, Shanghai Institute of Plant Physiology and Ecology, Chinese Academy of Sciences, Shanghai 200032, China; (J.H.); (B.Y.)
| | - Yang Li
- Microbial Research Institute of Liaoning Province, Chaoyang 122000, China; (Y.L.); (Y.G.); (C.X.); (F.C.); (J.C.)
| | - Bing Yang
- Laboratory of Photosynthesis and Environment, CAS Center for Excellence in Molecular Plant Sciences, Shanghai Institute of Plant Physiology and Ecology, Chinese Academy of Sciences, Shanghai 200032, China; (J.H.); (B.Y.)
| | - Yanli Guan
- Microbial Research Institute of Liaoning Province, Chaoyang 122000, China; (Y.L.); (Y.G.); (C.X.); (F.C.); (J.C.)
| | - Chong Xu
- Microbial Research Institute of Liaoning Province, Chaoyang 122000, China; (Y.L.); (Y.G.); (C.X.); (F.C.); (J.C.)
| | - Fei Chen
- Microbial Research Institute of Liaoning Province, Chaoyang 122000, China; (Y.L.); (Y.G.); (C.X.); (F.C.); (J.C.)
| | - Jingliang Chi
- Microbial Research Institute of Liaoning Province, Chaoyang 122000, China; (Y.L.); (Y.G.); (C.X.); (F.C.); (J.C.)
| | - Yongming Bao
- School of Bioengineering, Dalian University of Technology, Dalian 116024, China;
- School of Food and Environmental Science and Technology, Dalian University of Technology, Panjin 12421, China
- Correspondence: ; Tel.: +86-411-8470-6344; Fax: +86-411-8470-6365
| |
Collapse
|
2
|
Abstract
This review provides an overview on components of the sphingolipid superfamily, on their localization and metabolism. Information about the sphingolipid biological activity in cell physiopathology is given. Recent studies highlight the role of sphingolipids in inflammatory process. We summarize the emerging data that support the different roles of the sphingolipid members in specific phases of inflammation: (1) migration of immune cells, (2) recognition of exogenous agents, and (3) activation/differentiation of immune cells.
Collapse
|
3
|
Usatyuk PV, He D, Bindokas V, Gorshkova IA, Berdyshev EV, Garcia JGN, Natarajan V. Photolysis of caged sphingosine-1-phosphate induces barrier enhancement and intracellular activation of lung endothelial cell signaling pathways. Am J Physiol Lung Cell Mol Physiol 2011; 300:L840-50. [PMID: 21478254 DOI: 10.1152/ajplung.00404.2010] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Sphingosine-1-phosphate (S1P) is a bioactive sphingolipid that mediates cellular functions by ligation via G protein-coupled S1P receptors. In addition to its extracellular action, S1P also has intracellular effects; however, the signaling pathways modulated by intracellular S1P remain poorly defined. We have previously demonstrated a novel pathway of intracellular S1P generation in human lung endothelial cells (ECs). In the present study, we examined the role of intracellular S1P generated by photolysis of caged S1P on EC barrier regulation and signal transduction. Intracellular S1P released from caged S1P caused mobilization of intracellular calcium, induced activation of MAPKs, redistributed cortactin, vascular endothelial cadherin, and β-catenin to cell periphery, and tightened endothelial barrier in human pulmonary artery ECs. Treatment of cells with pertussis toxin (PTx) had no effect on caged S1P-mediated effects on Ca(2+) mobilization, reorganization of cytoskeleton, cell adherens junction proteins, and barrier enhancement; however, extracellular S1P effects were significantly attenuated by PTx. Additionally, intracellular S1P also activated small GTPase Rac1 and its effector Ras GTPase-activating-like protein IQGAP1, suggesting involvement of these proteins in the S1P-mediated changes in cell-to-cell adhesion contacts. Downregulation of sphingosine kinase 1 (SphK1), but not SphK2, with siRNA or inhibition of SphK activity with an inhibitor 2-(p-hydroxyanilino)-4-(p-chlorophenyl) thiazole (CII) attenuated exogenously administrated S1P-induced EC permeability. Furthermore, S1P1 receptor inhibitor SB649164 abolished exogenous S1P-induced transendothelial resistance changes but had no effect on intracellular S1P generated by photolysis of caged S1P. These results provide evidence that intracellular S1P modulates signal transduction in lung ECs via signaling pathway(s) independent of S1P receptors.
Collapse
Affiliation(s)
- Peter V Usatyuk
- Department of Pharmacology, University of Illinois at Chicago, 60612, USA
| | | | | | | | | | | | | |
Collapse
|
4
|
Basu A, Mazor S, Casey JR. Distance Measurements within a Concatamer of the Plasma Membrane Cl−/HCO3− Exchanger, AE1. Biochemistry 2010; 49:9226-40. [DOI: 10.1021/bi101134h] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Affiliation(s)
- Arghya Basu
- Membrane Protein Research Group, Department of Physiology and Department of Biochemistry, School of Molecular and Systems Medicine, 721 Medical Sciences Building, University of Alberta, Edmonton, Alberta, Canada T6G 2H7
| | - Shirley Mazor
- Membrane Protein Research Group, Department of Physiology and Department of Biochemistry, School of Molecular and Systems Medicine, 721 Medical Sciences Building, University of Alberta, Edmonton, Alberta, Canada T6G 2H7
| | - Joseph R. Casey
- Membrane Protein Research Group, Department of Physiology and Department of Biochemistry, School of Molecular and Systems Medicine, 721 Medical Sciences Building, University of Alberta, Edmonton, Alberta, Canada T6G 2H7
| |
Collapse
|
5
|
Abstract
Phagocytosis is an important component of innate and adaptive immunity. The formation of phagosomes and the subsequent maturation that capacitates them for pathogen elimination and antigen presentation are complex processes that involve signal transduction, cytoskeletal reorganization, and membrane remodeling. Lipids are increasingly appreciated to play a crucial role in these events. Sphingolipids, cholesterol, and glycerophospholipids, notably the phosphoinositides, are required for the segregation of signaling microdomains and for the generation of second messengers. They are also instrumental in the remodeling of the actin cytoskeleton and in directing membrane traffic. They accomplish these feats by congregating into liquid-ordered domains, by generating active metabolites that activate receptors, and by recruiting and anchoring specific protein ligands to the membrane, often altering their conformation and catalytic activity. A less appreciated role of acidic phospholipids is their contribution to the negative surface charge of the inner leaflet of the plasmalemma. The unique negativity of the inner aspect of the plasma membrane serves to attract and anchor key signaling and effector molecules that are required to initiate phagosome formation. Conversely, the loss of charge that accompanies phospholipid metabolism as phagosomes seal facilitates the dissociation of proteins and the termination of signaling and cytoskeleton assembly. In this manner, lipids provide a binary electrostatic switch to control phagocytosis.
Collapse
Affiliation(s)
- Tony Yeung
- Cell Biology Program, Hospital for Sick Children, Toronto, Ontario, Canada
| | | |
Collapse
|
6
|
Afrasiabi E, Blom T, Ekokoski E, Tuominen RK, Törnquist K. Sphingosylphosphorylcholine enhances calcium entry in thyroid FRO cells by a mechanism dependent on protein kinase C. Cell Signal 2006; 18:1671-8. [PMID: 16490345 DOI: 10.1016/j.cellsig.2006.01.005] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2005] [Accepted: 01/11/2006] [Indexed: 11/23/2022]
Abstract
Several sphingolipid derivatives, including sphingosylphosphorylcholine (SPC), regulate a multitude of biological processes. In the present study we show that both human thyroid cancer cells (FRO cells) and normal human thyroid cells express G protein-coupled receptor 4 (GPR4) and ovarian cancer G protein-coupled receptor 1 (OGR1), putative SPC-specific receptors. In FRO cells SPC evoked a concentration-dependent increase in intracellular free calcium concentration ([Ca2+]i) in a calcium containing, but not in a calcium-free buffer. Sphingosine 1-phosphate (S1P) evoked an increase in [Ca2+]i in both a calcium containing and a calcium-free buffer. The phospholipase C (PLC) inhibitor U 73122 potently attenuated the effect of SPC, suggesting that effects of SPC were mediated by a G protein coupled receptor. Overnight pretreatment of the cells with pertussis toxin did not affect the SPC-evoked response. Interestingly, SPC did not evoke an increase in inositol phosphates, although S1P did so. Furthermore, in cells pretreated with thapsigargin to deplete intracellular calcium stores, SPC still evoked an increase in [Ca2+]i, suggesting that SPC mainly evoked entry of extracellular calcium. When the cells were pretreated with the protein kinase C (PKC) inhibitor GF 109203X, or when the cells were pretreated with PMA for 24 h, the SPC-evoked calcium entry was attenuated. Thus, the SPC-evoked calcium entry was apparently dependent on PKC. In sharp contrast, the increase in [Ca2+]i evoked by S1P was not sensitive to GF 109203X. Furthermore, the calcium entry evoked by the diacylglycerol analog 1-oleoyl-2-acetyl-sn-glycerol was not inhibited by GF 109203X. In addition, SPC decreased the incorporation of 3H-thymidine in a concentration-dependent manner in FRO cells. Taken together, SPC may be an important factor regulating thyroid cancer cell function.
Collapse
Affiliation(s)
- Emad Afrasiabi
- Department of Biology, Abo Akademi University, BioCity, Artillerigatan 6, 20520 Turku, Finland
| | | | | | | | | |
Collapse
|
7
|
Lee HS, Park CS, Lee YM, Suk HY, Clemons TCM, Choi OH. Antigen-induced Ca2+ mobilization in RBL-2H3 cells: Role of I(1,4,5)P3 and S1P and necessity of I(1,4,5)P3 production. Cell Calcium 2005; 38:581-92. [PMID: 16219349 DOI: 10.1016/j.ceca.2005.08.002] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2005] [Revised: 08/30/2005] [Accepted: 08/30/2005] [Indexed: 01/08/2023]
Abstract
Inositol 1,4,5-trisphosphate (IP3) has long been recognized as a second messenger for intracellular Ca2+ mobilization. Recently, sphingosine 1-phosphate (S1P) has been shown to be involved in Ca2+ release from the endoplasmic reticulum (ER). Here, we investigated the role of S1P and IP3 in antigen (Ag)-induced intracellular Ca2+ mobilization in RBL-2H3 mast cells. Antigen-induced intracellular Ca2+ mobilization was only partially inhibited by the sphingosine kinase inhibitor dl-threo-dihydrosphingosine (DHS) or the IP3 receptor inhibitor 2-aminoethoxydiphenyl borate (2-APB), whereas preincubation with both inhibitors led to complete inhibition. In contrast, stimulation of A3 adenosine receptors with N5-ethylcarboxamidoadenosine (NECA) caused intracellular Ca2+ mobilization that was completely abolished by 2-APB but not by DHS, suggesting that NECA required only the IP3 pathway, while antigen used both the IP3 and S1P pathways. Interestingly, however, inhibition of IP3 production with the phospholipase C inhibitor U73122 completely abolished Ca2+ release from the ER induced by either stimulant. This suggested that S1P alone, without concomitant production of IP3, would not cause intracellular Ca2+ mobilization. This was further demonstrated in some clones of RBL-2H3 cells excessively overexpressing a beta isoform of Class II phosphatidylinositol 3-kinase (PI3KC2beta). In such clones including clone 5A4C, PI3KC2beta was overexpressed throughout the cell, although endogenous PI3KC2beta was normally expressed only in the ER. Overexpression of PI3KC2beta in the cytosol and the PM led to depletion of phosphatidylinositol 4,5-bisphosphate (PI(4,5)P2), resulting in a marked reduction in IP3 production. This could explain the abolishment of intracellular Ca2+ mobilization in clone 5A4C. Supporting this hypothesis, the Ca2+ mobilization was reconstituted by the addition of exogenous PI(4,5)P2 in these cells. Our results suggest that both IP3 and S1P contribute to FcvarepsilonRI-induced Ca2+ release from the ER and production of IP3 is necessary for S1P to cause Ca2+ mobilization from the ER.
Collapse
Affiliation(s)
- Hyun-Sil Lee
- Department of Medicine, Division of Allergy and Clinical Immunology, the Johns Hopkins University School of Medicine, JHAAC, Room 2A44a, 5501 Hopkins Bayview Circle, Baltimore, MD 21224, USA
| | | | | | | | | | | |
Collapse
|
8
|
Cavalli AL, O'Brien NW, Barlow SB, Betto R, Glembotski CC, Palade PT, Sabbadini RA. Expression and functional characterization of SCaMPER: a sphingolipid-modulated calcium channel of cardiomyocytes. Am J Physiol Cell Physiol 2003; 284:C780-90. [PMID: 12421694 DOI: 10.1152/ajpcell.00382.2002] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Calcium channels are important in a variety of cellular events including muscle contraction, signaling, proliferation, and apoptosis. Sphingolipids have been recognized as mediators of intracellular calcium release through their actions on a calcium channel, sphingolipid calcium release-mediating protein of the endoplasmic reticulum (SCaMPER). The current study investigates the expression and function of SCaMPER in cardiomyocytes. Northern analyses and RT-PCR cloning and sequencing revealed SCaMPER expression in both human and rat cardiac tissue. Immunofluorescence and Western blot analyses demonstrated that SCaMPER is abundant in cardiac tissue and is localized to the sarcotubular junction. This was confirmed by the colocalization of SCaMPER with dihydropyridine and ryanodine receptors by confocal microscopy. Purified T tubules were shown to contain SCaMPER and immunoelectron micrographs suggested that SCaMPER is located to the junctional T tubules, but a junctional SR localization cannot be ruled out. The sphingolipid ligand for SCaMPER, sphingosylphosphorylcholine (SPC), initiated calcium release from the cardiomyocyte SR. Importantly, antisense knockdown of SCaMPER mRNA produced a substantial reduction of sphingolipid-induced calcium release, suggesting that SCaMPER is a potentially important calcium channel of cardiomyocytes.
Collapse
MESH Headings
- Animals
- Animals, Newborn
- Calcium/metabolism
- Calcium Channels/genetics
- Calcium Channels/metabolism
- Calcium Signaling/genetics
- Cell Membrane/genetics
- Cell Membrane/metabolism
- DNA, Complementary/analysis
- DNA, Complementary/genetics
- Dimerization
- Immunohistochemistry
- Intracellular Membranes/metabolism
- Intracellular Membranes/ultrastructure
- Microscopy, Electron
- Microtubules/genetics
- Microtubules/metabolism
- Microtubules/ultrastructure
- Molecular Sequence Data
- Myocytes, Cardiac/metabolism
- Myocytes, Cardiac/ultrastructure
- Protein Structure, Quaternary/genetics
- RNA, Messenger/genetics
- RNA, Messenger/isolation & purification
- Rats
- Ryanodine Receptor Calcium Release Channel/metabolism
- Sarcoplasmic Reticulum/genetics
- Sarcoplasmic Reticulum/metabolism
- Sarcoplasmic Reticulum/ultrastructure
- Sequence Homology, Amino Acid
- Sequence Homology, Nucleic Acid
- Sphingolipids/metabolism
Collapse
Affiliation(s)
- Amy L Cavalli
- SDSU Heart Institute and Department of Biology, San Diego State University, California 92182-4614, USA
| | | | | | | | | | | | | |
Collapse
|
9
|
Abstract
This review presents the structural and functional role of phospholipids in chromatin and nuclear matrix as well as the difference in composition and turnover compared to those present in the nuclear membrane. Nuclei have a very active lipid metabolism which seems to play an important role in the transduction of the signals to the genome in response to agonists acting at the plasma membrane level. The evidence on the presence of phospholipid-calcium-dependent protein kinase C (PKC) in nuclei and enzymes of phospholipids turnover is given. Protein kinase C interacts with nuclear phosphoinositol and sphingomyelin cycles products. This fact evidences about possibility that signal transduction events could also occur at the nuclear level during induction of cell proliferation, differentiation and apoptosis.
Collapse
Affiliation(s)
- A V Alessenko
- Institute of Biochemical Physics of the Russian Academy of Sciences, Kosygin str, 4, 117977, Moscow, Russia.
| | | |
Collapse
|
10
|
Young KW, Nahorski SR. Intracellular sphingosine 1-phosphate production: a novel pathway for Ca2+ release. Semin Cell Dev Biol 2001; 12:19-25. [PMID: 11162743 DOI: 10.1006/scdb.2000.0213] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Sphingolipids such as sphingosine 1-phosphate (SPP) and sphingosylphosphorylcholine have long been recognized to possess Ca2+ mobilizing activity, yet to date little is known about their mechanism of action, or indeed their significance as Ca2+ mobilizing intracellular messengers. The recent discovery of extracellular receptors for the sphingolipids has further complicated the interpretation of many experiments in this field. This paper reviews the current literature in which molecular and pharmacological approaches have begun to uncover the signalling components associated with intracellular SPP production and Ca2+ mobilization. The functional significance of this novel Ca2+ release pathway is also discussed.
Collapse
Affiliation(s)
- K W Young
- Department of Cell Physiology and Pharmacology, Medical Sciences Building, University of Leicester, University Road, Leicester, LE1 9HN, UK.
| | | |
Collapse
|
11
|
Metzler DE, Metzler CM, Sauke DJ. Lipids, Membranes, and Cell Coats. Biochemistry 2001. [DOI: 10.1016/b978-012492543-4/50011-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
|
12
|
Young KW, Bootman MD, Channing DR, Lipp P, Maycox PR, Meakin J, Challiss RA, Nahorski SR. Lysophosphatidic acid-induced Ca2+ mobilization requires intracellular sphingosine 1-phosphate production. Potential involvement of endogenous EDG-4 receptors. J Biol Chem 2000; 275:38532-9. [PMID: 10954727 DOI: 10.1074/jbc.m006631200] [Citation(s) in RCA: 56] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Lysophosphatidic acid (LPA)-mediated Ca(2+) mobilization in human SH-SY5Y neuroblastoma cells does not involve either inositol 1,4, 5-trisphosphate (Ins(1,4,5)P(3))- or ryanodine-receptor pathways, but is sensitive to inhibitors of sphingosine kinase. This present study identifies Edg-4 as the receptor subtype involved and investigates the presence of a Ca(2+) signaling cascade based upon the lipid second messenger molecule, sphingosine 1-phosphate. Both LPA and direct G-protein activation increase [(3)H]sphingosine 1-phosphate levels in SH-SY5Y cells. Measurements of (45)Ca(2+) release in premeabilized SH-SY5Y cells indicates that sphingosine 1-phosphate, sphingosine, and sphingosylphosphorylcholine, but not N-acetylsphingosine are capable of mobilizing intracellular Ca(2+). Furthermore, the effect of sphingosine was attenuated by the sphingosine kinase inhibitor dimethylsphingosine, or removal of ATP. Confocal microscopy demonstrated that LPA stimulated intracellular Ca(2+) "puffs," which resulted from an interaction between the sphingolipid Ca(2+) release pathway and Ins(1,4,5)P(3) receptors. Down-regulation of Ins(1,4,5)P(3) receptors uncovered a Ca(2+) response to LPA, which was manifest as a progressive increase in global cellular Ca(2+) with no discernible foci. We suggest that activation of an LPA-sensitive Edg-4 receptor solely utilizes the production of intracellular sphingosine 1-phosphate to stimulate Ca(2+) mobilization in SH-SY5Y cells. Unlike traditional Ca(2+) release processes, this novel pathway does not require the progressive recruitment of elementary Ca(2+) events.
Collapse
MESH Headings
- Caffeine/pharmacology
- Calcium/metabolism
- Calcium Channels/physiology
- Calcium Signaling
- Guanosine 5'-O-(3-Thiotriphosphate)/metabolism
- Heterotrimeric GTP-Binding Proteins/metabolism
- Humans
- Inositol 1,4,5-Trisphosphate/metabolism
- Inositol 1,4,5-Trisphosphate Receptors
- Kinetics
- Lysophospholipids/pharmacology
- Neuroblastoma
- Receptors, Cell Surface/drug effects
- Receptors, Cell Surface/genetics
- Receptors, Cell Surface/physiology
- Receptors, Cytoplasmic and Nuclear/physiology
- Receptors, G-Protein-Coupled
- Receptors, Lysophosphatidic Acid
- Reverse Transcriptase Polymerase Chain Reaction
- Sphingosine/analogs & derivatives
- Sphingosine/metabolism
- Sphingosine/pharmacology
- Tumor Cells, Cultured
Collapse
Affiliation(s)
- K W Young
- Department of Cell Physiology and Pharmacology, Medical Sciences Building, University of Leicester, University Road, Leicester, LE1 9HN United Kingdom
| | | | | | | | | | | | | | | |
Collapse
|
13
|
Augé N, Nègre-Salvayre A, Salvayre R, Levade T. Sphingomyelin metabolites in vascular cell signaling and atherogenesis. Prog Lipid Res 2000; 39:207-29. [PMID: 10799716 DOI: 10.1016/s0163-7827(00)00007-2] [Citation(s) in RCA: 83] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
The atherosclerotic lesion most probably develops through a number of cellular events which implicate all vascular cell types and include synthesis of extracellular proteins, cell proliferation, differentiation and death. Sphingolipids and sphingolipid metabolizing enzymes may play important roles in atherogenesis, not only because of lipoprotein alterations but also by mediating a number of cellular events which are believed to be crucial in the development of the vascular lesions such as proliferation or cell death. Exogenous sphingolipids may mediate various biological effects such as apoptosis, mitogenesis or differentiation depending on the cell type. Moreover, several molecules present in the atherogenic lesion, such as oxidized LDL, growth factors or cytokines, which activate intracellular signaling pathways leading to vascular cell modifications, can stimulate sphingomyelin hydrolysis and generation of ceramide (and other metabolites as sphingosine-1-phosphate). Here we review the potential implication of the sphingomyelin/ceramide cycle in vascular cell signaling related to atherosclerosis, and more generally the role of sphingolipids in the events observed during the atherosclerotic process as cell differentiation, migration, adhesion, retraction, proliferation and death.
Collapse
Affiliation(s)
- N Augé
- Laboratoire de Biochimie, INSERM U. 466, "Maladies Métaboliques," Institut Louis Bugnard, Bât. Université Paul Sabatier, CHU Rangueil, 1 Avenue Jean Poulhès, F-31403, Toulouse, France.
| | | | | | | |
Collapse
|
14
|
Sharma C, Smith T, Li S, Schroepfer GJ, Needleman DH. Inhibition of Ca2+ release channel (ryanodine receptor) activity by sphingolipid bases: mechanism of action. Chem Phys Lipids 2000; 104:1-11. [PMID: 10660207 DOI: 10.1016/s0009-3084(99)00106-1] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Sphingosine inhibits the activity of the skeletal muscle Ca2+ release channel (ryanodine receptor) and is a noncompetitive inhibitor of [3H]ryanodine binding (Needleman et al., Am. J. Physiol. 272, C1465-1474, 1997). To determine the contribution of other sphingolipids to the regulation of ryanodine receptor activity, several sphingolipid bases were assessed for their ability to alter [3H]ryanodine binding to sarcoplasmic reticulum (SR) membranes and to modulate the activity of the Ca2+ release channel. Three lipids, N,N-dimethylsphingosine, dihydrosphingosine, and phytosphingosine, inhibited [3H]ryanodine binding to both skeletal and cardiac SR membranes. However, the potency of these three lipids and sphingosine was lower in rabbit cardiac membranes when compared to rabbit skeletal muscle membranes and when compared to sphingosine. Like sphingosine, the lipids inhibited [3H]ryanodine binding by greatly increasing the rate of dissociation of bound [3H]ryanodine from SR membranes, indicating that these three sphingolipid bases were noncompetitive inhibitors of [3H]ryanodine binding. These bases also decreased the activity of the Ca2+ release channel incorporated into planar lipid bilayers by stabilizing a long closed state. Sphingosine-1-PO4 and C6 to C18 ceramides of sphingosine had no significant effect on [3H]ryanodine binding to cardiac or skeletal muscle SR membranes. Saturation of the double bond at positions 4-5 decreased the ability of the sphingolipid bases to inhibit [3H]ryanodine binding 2-3 fold compared to sphingosine. In summary, our data indicate that other endogenous sphingolipid bases are capable of modulating the activity of the Ca2+ release channel and as a class possess a common mechanism of inhibition.
Collapse
Affiliation(s)
- C Sharma
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX 77030, USA
| | | | | | | | | |
Collapse
|
15
|
Uehara A, Yasukochi M, Imanaga I, Berlin JR. Effect of sphingosylphosphorylcholine on the single channel gating properties of the cardiac ryanodine receptor. FEBS Lett 1999; 460:467-71. [PMID: 10556518 DOI: 10.1016/s0014-5793(99)01385-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
The effects of the lysosphingolipid, sphingosylphosphorylcholine (SPC), on the cardiac ryanodine receptor (RyR) were examined. The open probability of cardiac RyR incorporated in lipid bilayers was decreased by cytoplasmic, but not lumenal side application of micromolar concentrations of SPC. Modification of channel function was characterized by the appearance of a long-lived closed state in addition to the brief channel closings observed in the presence and absence of SPC. Open channel kinetics and ion conduction properties, however, were not altered by this compound. These results suggest that SPC, a putative second messenger derived from sphingomyelin, may regulate Ca(2+) release from the sarcoplasmic reticulum by modifying the gating kinetics of the RyR.
Collapse
Affiliation(s)
- A Uehara
- Department of Physiology, School of Medicine, Fukuoka University, 45-1, 7-chome Nanakuma, Jonan-ku, Fukuoka, Japan.
| | | | | | | |
Collapse
|
16
|
Parris JR, Cobban HJ, Littlejohn AF, MacEwan DJ, Nixon GF. Tumour necrosis factor-alpha activates a calcium sensitization pathway in guinea-pig bronchial smooth muscle. J Physiol 1999; 518 ( Pt 2):561-9. [PMID: 10381600 PMCID: PMC2269439 DOI: 10.1111/j.1469-7793.1999.0561p.x] [Citation(s) in RCA: 64] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
1. The effects of tumour necrosis factor-alpha (TNF) on guinea-pig bronchial smooth muscle contractility were investigated. 2. The Ca2+-activated contractile response of permeabilized bronchial smooth muscle strips was significantly increased after incubation with 1 microgram ml-1 TNF for 45 min. This TNF-induced effect was not due to a further increase in intracellular Ca2+. 3. The TNF-induced Ca2+ sensitization was, at least partly, the result of an increase in myosin light chain20 phosphorylation. 4. The intracellular signalling pathway involved in this effect of TNF was further investigated. Sphingomyelinase, a potential mediator of TNF, had no effect on Ca2+ sensitivity of permeabilized bronchial smooth muscle. Also, p42/p44 mitogen-activated protein kinase (p42/p44mapk), activated by TNF in some cell types, did not show an increased activation in bronchial smooth muscle after TNF treatment. 5. In conclusion, TNF may activate a novel signalling pathway in guinea-pig bronchial smooth muscle leading to an increase in myosin light chain20 phosphorylation and a subsequent increase in Ca2+ sensitivity of the myofilaments. This pathway does not appear to involve sphingomyelinase-liberated ceramides or activation of p42/p44mapk. Given the importance of TNF in asthma, this TNF-induced Ca2+ sensitization of the myofilaments may represent a mechanism responsible for airway hyper-responsiveness.
Collapse
Affiliation(s)
- J R Parris
- Department of Biomedical Sciences, Institute of Medical Sciences, University of Aberdeen, Aberdeen, UK
| | | | | | | | | |
Collapse
|
17
|
Abstract
The high affinity receptor for immunoglobulin E (designated Fc epsilon RI) is the member of the antigen (Ag) receptor superfamily responsible for linking pathogen-or allergen-specific IgEs with cellular immunologic effector functions. This review provides background information on Fc epsilon RI function combined with more detailed summaries of recent progress in understanding specific aspects of Fc epsilon RI biology and biochemistry. Topics covered include the coordination and function of the large multiprotein signaling complexes that are assembled when Fc epsilon RI and other Ag receptors are engaged, new information on human receptor structures and tissue distribution, and the role of the FcR beta chain in signaling and its potential contribution to atopic phenotypes.
Collapse
Affiliation(s)
- J P Kinet
- Harvard Medical School, Boston, Massachusetts, USA.
| |
Collapse
|
18
|
Schick A, Schwarzmann G, Kolter T, Sandhoff K. Synthesis of tritium labelled phosphonate analogues of sphinganine-1-phosphate. J Labelled Comp Radiopharm 1998. [DOI: 10.1002/(sici)1099-1344(199705)39:5<441::aid-jlcr988>3.0.co;2-d] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
19
|
Orlati S, Porcelli AM, Hrelia S, Lorenzini A, Rugolo M. Intracellular calcium mobilization and phospholipid degradation in sphingosylphosphorylcholine-stimulated human airway epithelial cells. Biochem J 1998; 334 ( Pt 3):641-9. [PMID: 9729473 PMCID: PMC1219734 DOI: 10.1042/bj3340641] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Extracellular sphingosylphosphorylcholine (SPC) caused a remarkable elevation in the intracellular Ca2+ concentration ([Ca2+]i) in immortalized human airway epithelial cells (CFNP9o-). An increase in total inositol phosphates formation was determined; however, the dose responses for [Ca2+]i elevation and inositol phosphates production were slightly different and, furthermore, PMA and pertussis toxin almost completely inhibited [Ca2+]i mobilization by SPC, whereas inositol phosphates production was only partially reduced. The possible direct interaction of SPC with Ca2+ channels of intracellular stores was determined by experiments with permeabilized cells, where SPC failed to evoke Ca2+ release, whereas lysophosphatidic acid was shown to be effective. The level of phosphatidic acid was increased by SPC only in the presence of AACOCF3, a specific inhibitor of phospholipase A2 (PLA2) and blocked by both pertussis toxin and R59022, an inhibitor of diacylglycerol kinase. R59022 enhanced diacylglycerol production by SPC and also significantly reduced [Ca2+]i mobilization. Only polyunsaturated diacylglycerol and phosphatidic acid were generated by SPC. Lastly, SPC caused stimulation of arachidonic acid release, indicating the involvement of PLA2. Taken together, these data suggest that, after SPC stimulation, phospholipase C-derived diacylglycerol is phosphorylated by a diacylglycerol kinase to phosphatidic acid, which is further hydrolysed by PLA2 activity to arachidonic and lysophosphatidic acids. We propose that lysophosphatidic acid might be the intracellular messenger able to release Ca2+ from internal stores.
Collapse
Affiliation(s)
- S Orlati
- Dip. di Biologia Ev. Sp., Università di Bologna, Bologna, Italy
| | | | | | | | | |
Collapse
|
20
|
van Echten-Deckert G, Schick A, Heinemann T, Schnieders B. Phosphorylated cis-4-methylsphingosine mimics the mitogenic effect of sphingosine-1-phosphate in Swiss 3T3 fibroblasts. J Biol Chem 1998; 273:23585-9. [PMID: 9722598 DOI: 10.1074/jbc.273.36.23585] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The phosphorylated derivative of sphingosine, sphingosine-1-phosphate, is a short-living metabolite of ultimate ceramide degradation and was shown to act as an intracellular signaling molecule, stimulating cell proliferation in quiescent Swiss 3T3 fibroblasts and inducing the release of calcium from intracellular stores (Zhang, H., Desai, N. N., Olivera, A., Seki, T., Brooker, G., and Spiegel, S. (1991) J. Cell. Biol. 114, 155-167). In the present study, 24-h treatment of Swiss 3T3 fibroblasts with the synthetic sphingosine analogue cis-4-methylsphingosine resulted in proliferation of quiescent Swiss 3T3 fibroblasts that was 3-fold stronger than that of equimolar sphingosine-1-phosphate. The phosphorylated derivative of cis-4-methylsphingosine accumulated drastically in the cells. Simultaneous treatment with the sphingosine kinase inhibitor L-threo-sphinganine reduced both the amount of phosphorylated cis-4-methylsphingosine and cell proliferation induced by this compound by about 50%, indicating that the phosphorylated derivative mediated the proliferative stimulus. The mitogenic effect of cis-4-methylsphingosine was associated with a mobilization of intracellular calcium in Swiss 3T3 fibroblasts that was similar to that induced by sphingosine-1-phosphate. The results demonstrate that the phosphorylated derivative of cis-4-methylsphingosine mimics the previously reported mitogenic action of sphingosine-1-phosphate in Swiss 3T3 cells, and the stronger effect most likely corresponds to the unusual accumulation of this compound.
Collapse
Affiliation(s)
- G van Echten-Deckert
- Kekulé-Institut für Organische Chemie und Biochemie der Universität Bonn, 53121 Bonn, Germany.
| | | | | | | |
Collapse
|
21
|
Meyer zu Heringdorf D, Niederdräing N, Neumann E, Fröde R, Lass H, Van Koppen CJ, Jakobs KH. Discrimination between plasma membrane and intracellular target sites of sphingosylphosphorylcholine. Eur J Pharmacol 1998; 354:113-22. [PMID: 9726638 DOI: 10.1016/s0014-2999(98)00436-1] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
On the background of the emerging concept of G protein-coupled sphingolipid receptors, Ca2+ mobilization by sphingosylphosphorylcholine (SPPC) in intact cells and SPPC-induced Ca2+ release in permeabilized cells, both occurring at similar, micromolar concentrations, were characterized and compared. In intact human embryonic kidney (HEK-293) cells, SPPC rapidly increased [Ca2+]i by mobilization of Ca2+ from thapsigargin-sensitive stores. In saponin-permeabilized HEK-293 cells, SPPC released stored Ca2+, in a manner similar to but independent of inositol 1,4,5-trisphosphate. Only the action of SPPC on intact cells, but not that in permeabilized cells, was, at least in part, sensitive to pertussis toxin. In addition and most important, Ca2+ release by SPPC in permeabilized cells was not stereoselective, whereas in intact cells only the naturally occurring D-erythro-SPPC, but not L-threo-SPPC, increased [Ca2+]i. Stereoselectivity of SPPC-induced [Ca2+]i increase was also demonstrated in bovine aortic endothelial cells. In conclusion, Ca2+ mobilization by SPPC in intact cells is independent of the previously described SPPC-gated Ca2+ channel on endoplasmic reticulum but probably mediated by a membrane sphingolipid receptor. Thus, SPPC can regulate Ca2+ homeostasis by acting apparently at two cellular targets, which exhibit clearly distinct recognition patterns.
Collapse
|
22
|
Duan RD. Sphingomyelin hydrolysis in the gut and clinical implications in colorectal tumorigenesis and other gastrointestinal diseases. Scand J Gastroenterol 1998; 33:673-83. [PMID: 9712229 DOI: 10.1080/00365529850171594] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Affiliation(s)
- R D Duan
- Dept. of Cell Biology 1, University Hospital of Lund, Sweden
| |
Collapse
|
23
|
Clementi E. Role of nitric oxide and its intracellular signalling pathways in the control of Ca2+ homeostasis. Biochem Pharmacol 1998; 55:713-8. [PMID: 9586942 DOI: 10.1016/s0006-2952(97)00375-4] [Citation(s) in RCA: 85] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Ca2+, a primary regulator of physiological functions in all cells, is involved in a variety of intracellular signalling pathways; control of Ca2+ homeostasis is, therefore, a fundamental cell activity. To this end, cells have developed a variety of mechanisms to ensure the buffering of Ca2+, its influx and extrusion from the plasma membrane, and its release/accumulation within specific intracellular storage compartments. Over the last few years, evidence gathered from a number of cell systems has indicated that one of the key messengers governing the overall control of Ca2+ homeostasis is nitric oxide (NO), which may be produced intracellularly or may originate from neighboring cells. The aim of the present commentary is to concentrate on the biochemical steps in Ca2+ homeostasis that are controlled by NO and to describe what is known thus far concerning the molecular mechanisms of its action. Particular attention will be given to the effects of NO on: (i) inositol 1,4,5-trisphosphate and cyclic ADP ribose generation; (ii) Ca2+ release from both inositol 1,4,5-trisphosphate-sensitive and ryanodine-sensitive Ca2+ stores; and (iii) Ca2+ influx via both store- and second messenger-operated Ca2+ channels. The evidence discussed here documents the complexity of the interactions between the Ca2+ and the NO signalling systems, which represent an extraordinary example of cross-talk operating at multiple sites and which are continuously active in the regulation of cytosolic Ca2+ (and NO) levels.
Collapse
Affiliation(s)
- E Clementi
- Department of Pharmacology, Faculty of Pharmacy, CNR-IBAF, University of Reggio Calabria, Catanzaro, Italy.
| |
Collapse
|
24
|
Berger A, Cultaro CM, Segal S, Spiegel S. The potent lipid mitogen sphingosylphosphocholine activates the DNA binding activity of upstream stimulating factor (USF), a basic helix-loop-helix-zipper protein. BIOCHIMICA ET BIOPHYSICA ACTA 1998; 1390:225-36. [PMID: 9507145 DOI: 10.1016/s0005-2760(97)00180-x] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
We previously demonstrated that the sphingolipid, sphingosylphosphocholine (SPC) increased DNA binding activity of AP-1 proteins accompanying cellular proliferation. Herein, the effects of SPC on DNA binding activity and transcription of the basic, helix-loop-helix, leucine zipper (bHLH-ZIP) proteins Myc, Max, and USF were investigated because they regulate genes involved in mitogenesis. E-box (CACGTG) DNA binding proteins were detected by electrophoretic mobility shift assays in nuclear extracts from Swiss 3T3 fibroblasts. The slowest migrating complex (complex I) increased within 1-3 min after treatment with SPC, remained elevated for 10 min, and increased again after 12 h. Complexes I and II contained USF-1 and USF-2 proteins, and complex I migrated similarly to recombinant USF-1 protein/DNA complex. Treatment of nuclear extracts with alkaline phosphatase decreased these complexes suggesting USF might be a phosphoprotein, post-translationally modified by SPC. max and usf-1 mRNA levels were unaffected by SPC treatment. In contrast, c-myc mRNA was rapidly elevated, reached maximum levels at 0.5-1 h, and showed an additional increase after 12 h, just preceding S phase. Thus, certain bHLH-ZIP transcription factors may be involved in cell growth regulation by SPC.
Collapse
Affiliation(s)
- A Berger
- Department of Biochemistry and Molecular Biology, Georgetown University Medical Center, Washington, DC 20007, USA
| | | | | | | |
Collapse
|
25
|
Titievsky A, Titievskaya I, Pasternack M, Kaila K, Törnquist K. Sphingosine inhibits voltage-operated calcium channels in GH4C1 cells. J Biol Chem 1998; 273:242-7. [PMID: 9417071 DOI: 10.1074/jbc.273.1.242] [Citation(s) in RCA: 41] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
In the present study we investigated the mechanism of inhibitory action of sphingosine (SP) on voltage-activated calcium channels (VOCCs) in pituitary GH4C1 cells. Using the patch-clamp technique in the whole-cell mode, we show that SP inhibits Ba2+ currents (IBa) when 0.1 mM BAPTA is included in the patch pipette. However, when the BAPTA concentration was raised to 1-10 mM, SP was without a significant effect. The effect of SP was apparently not mediated via a kinase, as it was not inhibited by staurosporine. By using the double-pulse protocol (to release possible functional inhibition of the VOCCs by G proteins), we observed that G proteins apparently evoked very little functional inhibition of the VOCCs. Furthermore, including GDPbetaS (guanyl-5'-yl thiophosphate) in the patch pipette did not alter the inhibitory effect of SP on the Ba2+ current, suggesting that SP did not modulate the VOCCs via a G protein-dependent pathway. Single-channel experiments with SP in the pipette, and experiments with excised outside-out patches, suggested that SP directly inhibited VOCCs. The main mechanism of action was a dose-dependent prolongation of the closed time of the channels. The results thus show that SP is a potent inhibitor of VOCCs in GH4C1 cells, and that calcium may be a cofactor in this inhibition.
Collapse
Affiliation(s)
- A Titievsky
- Department of Biosciences, Division of Animal Physiology, University of Helsinki, Helsinki, and the Department of Biology, Abo Akademi University, Turku, Finland
| | | | | | | | | |
Collapse
|
26
|
Sciorati C, Rovere P, Ferrarini M, Heltai S, Manfredi AA, Clementi E. Autocrine nitric oxide modulates CD95-induced apoptosis in gammadelta T lymphocytes. J Biol Chem 1997; 272:23211-5. [PMID: 9287328 DOI: 10.1074/jbc.272.37.23211] [Citation(s) in RCA: 89] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Gammadelta T lymphocytes play an important early role in the defense against pathogens. Their function is terminated by acquisition of susceptibility to CD95-triggered apoptosis. Here we show that the regulation of this process depends on the activity of the endothelial NO synthase expressed by gammadelta T lymphocytes, which is modulated in an activation-dependent way. The effects of nitric oxide thus generated, mediated via cGMP generation, are exerted at at least two sites along the CD95 signaling cascade: one at, or upstream, and the other downstream of ceramide generation. At either site, nitric oxide/cGMP action is sufficient for protection from apoptosis. The effect of NO is selective for apoptosis induced by CD95 cross-linking, since it does not affect apoptotic program triggered by other stimuli. The evidence here reported demonstrates a new physiological role for nitric oxide, acting as a survival factor for T lymphocytes.
Collapse
Affiliation(s)
- C Sciorati
- Receptor Biochemistry Unit, DIBIT, University of Reggio Calabria, 88021 Catanzaro and Consiglio Nazionale delle Ricerche Cellular and Molecular Pharmacology Centre, Via Olgettina 58, 20132 Milano, Italy
| | | | | | | | | | | |
Collapse
|
27
|
Törnquist K, Woodside M, Grinstein S. Sphingosylphosphorylcholine activates an amiloride-insensitive Na+-H+-exchange mechanism in GH4C1 cells. EUROPEAN JOURNAL OF BIOCHEMISTRY 1997; 248:394-400. [PMID: 9346294 DOI: 10.1111/j.1432-1033.1997.00394.x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
The effect of sphingosylphosphorylcholine (SphPCho) on the intracellular pH (pHi) in GH4C1 cells was investigated. SphPCho evoked a very slow increase in basal pHi. In cells acidified with nigericin, SphPCho induced a rapid alkalinization of the cells. The effect was inhibited in a Na+-free buffer solution, but was insensitive to ethylisopropyl amiloride, a potent inhibitor of Na+-H+ exchangers (NHE). Reverse transcription and PCR showed that the predominant isoform of the antiport expressed in GH4C1 cells is NHE-1. The rate of alkalinization after stimulation with propionate, and after addition of Na+ to cells acidified with NH4Cl, was enhanced in cells treated with SphPCho. The initial rate of alkalinization after addition of Na+ to acidified cells treated with SphPCho gave an apparent Km value of 15 +/- 2 mM for Na+. The Vmax value was 9 +/- 2 mM H+/min. The effect was insensitive to ouabain, staurosporine and bafilomycin A. However, the SphPCho-evoked alkalinization was abolished in cells treated with 2-deoxy-D-glucose. The effect was not due to the charge of the molecule, as stearylamine increased pHi in Na+-containing and Na+-free buffer. The results show that SphPCho may activate Na+-H+ exchange, and that this effect is mediated via an amiloride-insensitive exchange mechanism.
Collapse
Affiliation(s)
- K Törnquist
- Department of Biosciences, University of Helsinki, Finland
| | | | | |
Collapse
|
28
|
Abstract
Sphingolipid breakdown products are now being recognized to play a dual role in cellular signalling, acting as intracellular as well as extracellular signalling molecules. Both types of action may even be found with one sphingolipid species. The recent demonstration of G protein-coupled receptors with high affinity for sphingosine 1-phosphate and sphingosylphosphorylcholine has been followed by the discovery of several novel sphingolipid actions, such as regulation of heart rate, oxidative burst, neurite retraction or platelet activation. Ligand profiles and concentration-response relationships suggest the existence of putative sphingolipid receptor subtypes. Against this background, several observations on supposed sphingolipid second messenger actions deserve a new evaluation.
Collapse
|
29
|
Needleman DH, Aghdasi B, Seryshev AB, Schroepfer GJ, Hamilton SL. Modulation of skeletal muscle Ca2(+)-release channel activity by sphingosine. THE AMERICAN JOURNAL OF PHYSIOLOGY 1997; 272:C1465-74. [PMID: 9176136 DOI: 10.1152/ajpcell.1997.272.5.c1465] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
The effect of D-erythro-C18-sphingosine (sphingosine) and related compounds on the Ca(2+)-release channel (ryanodine binding protein) was examined on rabbit skeletal muscle membranes, on the purified ryanodine binding protein, and on the channel reconstituted into planar lipid bilayers. Sphingosine inhibited [3H]ryanodine binding to sarcoplasmic reticulum (SR) membranes in a dose-dependent manner similar to published results (R. A. Sabbadini, R. Betto, A. Teresi, G. Fachechi-Cassano, and G. Salviati. J. Biol. Chem. 267: 15475-15484, 1992). The sphingolipid also inhibited [3H]ryanodine binding to the purified ryanodine binding protein. Our results demonstrate that the inhibition of [3H]ryanodine binding by sphingosine is due to an increased rate of dissociation of bound [3H]ryanodine from SR membranes and a decreased rate of association of [3H]ryanodine to the high-affinity site. Unlike other modulators of the Ca(2+)-release channel, sphingosine can remove bound [3H]ryanodine from the high-affinity site within minutes. Sphingosine increased the rate of dissociation of [3H]ryanodine bound to a solubilized proteolytic fragment derived from the carboxy terminus of the ryanodine binding protein (cleavage at Arg4475). Sphingosine also inhibited the activity of the Ca(2+)-release channel incorporated into planar lipid bilayers. Taken together, the data provide evidence for a direct effect of sphingosine on the Ca(2+)-release channel. Sphingosine is a noncompetitive inhibitor at the high-affinity ryanodine binding site, and it interacts with a site between Arg4475 and the carboxy terminus of the Ca(2+)-release channel.
Collapse
Affiliation(s)
- D H Needleman
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, Texas 77030, USA
| | | | | | | | | |
Collapse
|
30
|
Betto R, Teresi A, Turcato F, Salviati G, Sabbadini RA, Krown K, Glembotski CC, Kindman LA, Dettbarn C, Pereon Y, Yasui K, Palade PT. Sphingosylphosphocholine modulates the ryanodine receptor/calcium-release channel of cardiac sarcoplasmic reticulum membranes. Biochem J 1997; 322 ( Pt 1):327-33. [PMID: 9078280 PMCID: PMC1218195 DOI: 10.1042/bj3220327] [Citation(s) in RCA: 39] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Sphingosylphosphocholine (SPC) modulates Ca2+ release from isolated cardiac sarcoplasmic reticulum membranes; 50 microM SPC induces the release of 70 80% of the accumulated calcium. SPC release calcium from cardiac sarcoplasmic reticulum through the ryanodine receptor, since the release is inhibited by the ryanodine receptor channel antagonists ryanodine. Ruthenium Red and sphingosine. In intact cardiac myocytes, even in the absence of extracellular calcium. SPC causes a rise in diastolic Ca2+, which is greatly reduced when the sarcoplasmic reticulum is depleted of Ca2+ by prior thapsigargin treatment. SPC action on the ryanodine receptor is Ca(2+)-dependent. SPC shifts to the left the Ca(2+)-dependence of [3H]ryanodine binding, but only at high pCa values, suggesting that SPC might increase the sensitivity to calcium of the Ca(2+)-induced Ca(2+)-release mechanism. At high calcium concentrations (pCa 4.0 or lower), where [3H]ryanodine binding is maximally stimulated, no effect of SPC is observed. We conclude that SPC releases calcium from cardiac sarcoplasmic reticulum membranes by activating the ryanodine receptor and possibly another intracellular Ca(2+)-release channel, the sphingolipid Ca(2+)-release-mediating protein of endoplasmic reticulum (SCaMPER) [Mao, Kim, Almenoff, Rudner, Kearney and Kindman (1996) Proc.Natl.Acad.Sci. U.S.A 93, 1993-1996], which we have identified for the first time in cardiac tissue.
Collapse
Affiliation(s)
- R Betto
- Department of Biomedical and Experimental Sciences, University of Padova, Italy
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Meyer zu Heringdrof D, van Koppen CJ, Windorfer B, Himmel HM, Jakobs KH. Calcium signalling by G protein-coupled sphingolipid receptors in bovine aortic endothelial cells. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 1996; 354:397-403. [PMID: 8897440 DOI: 10.1007/bf00168428] [Citation(s) in RCA: 42] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Besides its role as a putative second messenger releasing Ca2+ from intracellular stores, sphingosine-1-phosphate (SPP) has recently been identified as an extracellularly acting ligand activating a high affinity G protein-coupled membrane receptor in various cell types. Since SPP can be released from activated platelets, we examined in the present study whether endothelial cells express receptors for SPP and related sphingolipids. In bovine aortic endothelial cells loaded with fura-2, addition of SPP caused a rapid and transient increase in intracellular Ca2+ concentration ([Ca2+]i), amounting to maximally about 230 nM. Removal of extracellular Ca2+ revealed that SPP-induced [Ca2+]i elevations were due to both release of Ca2+ from intracellular stores and influx of extracellular Ca2+. Pretreatment of the cells with pertussis toxin inhibited the SPP-induced increase in [Ca2+]i by 83%, in line with the previously reported involvement of G proteins of the Gi/o family in SPP signalling in other cell types. In contrast to other [Ca2+]i-elevating agonists, e.g., ATP and bradykinin, SPP did not activate phospholipase C in bovine aortic endothelial cells, suggesting the involvement of a novel, unidentified signalling pathway in SPP-induced release of intracellular Ca2+. Furthermore, SPP also did not cause activation of either phospholipase D or A2. Out of various related sphingolipids studied, only sphingosylphosphorylcholine (SPPC) induced a similar maximal increase in [Ca2+]i as SPP, and its effect was also fully pertussis toxin-sensitive. However, the potencies of the two sphingolipids to increase [Ca2+]i differed by more than two orders of magnitude, with the EC50 values being 0.8 nM and 260 nM for SPP and SPPC, respectively. These results identify SPP and SPPC as novel and potent endothelial agonists, inducing calcium signalling by activation of a Gi/o protein-coupled receptor(s). Given the recently reported release of SPP from thrombin-activated platelets, SPP may represent a novel mediator of platelet-endothelial cell interactions.
Collapse
|
32
|
Sakano S, Takemura H, Yamada K, Imoto K, Kaneko M, Ohshika H. Ca2+ mobilizing action of sphingosine in Jurkat human leukemia T cells. Evidence that sphingosine releases Ca2+ from inositol trisphosphate- and phosphatidic acid-sensitive intracellular stores through a mechanism independent of inositol trisphosphate. J Biol Chem 1996; 271:11148-55. [PMID: 8626660 DOI: 10.1074/jbc.271.19.11148] [Citation(s) in RCA: 42] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
Effects of sphingosine on Ca2+ mobilization in the human Jurkat T cell line were examined. Sphingosine increased the cytoplasmic Ca2+ concentration ([Ca2+]i) in a dose-dependent manner with an ED50 of around 8 microM. Sphingosine and OKT3, a CD3 monoclonal antibody, transiently increased [Ca2+]i, which declined to the resting level in the absence of extracellular Ca2+. Under the same conditions, pretreatment with sphingosine inhibited but did not abolish an increase in [Ca2+]i induced by the subsequent addition of OKT3 and vice versa. However, pretreatment with sphingosine did not affect an increase in [Ca2+]i induced by OKT3 in the presence of Ca2+. OKT3 increased IP3 formation, but sphingosine did not affect the level of IP3 by itself nor did it cause IP3 formation induced by OKT3. In permeabilized Jurkat cells, the addition of IP3 released Ca2+ from nonmitochondrial intracellular stores, but the addition of sphingosine did not. Sphingosine, stearylamine, and psychosine increased [Ca2+]i and diacylglycerol (DG) kinase activation; however, ceramide did not, whereas sphingosine 1-phosphate slightly activated DG kinase without elevation of [Ca2+]i. Pretreatment with R59022, a DG kinase inhibitor, abolished the peak but did not affect the sustained response to [Ca2+]i to sphingosine. Phosphatidic acid (PA) elevated [Ca2+]i, after which it declined to a resting level even in the presence of extracellular Ca2+. In accordance with this, PA did not stimulate 45Ca2+ uptake into cells, but sphingosine and OKT3 did. Pretreatment with PA partially inhibited a rise in [Ca2+]i induced by the subsequent addition of sphingosine and vice versa in the absence of extracellular Ca2+. Under similar conditions, pretreatment with PA affected an elevation of [Ca2+]i induced by OKT3 less, after which the subsequent addition of sphingosine did not increase [Ca2+]i. In permeabilized Jurkat cells, the addition of IP3 did not release Ca2+, but PA did in the presence of heparin. Pretreatment with thapsigargin, a microsomal Ca2+-ATPase inhibitor, abolished the rises of [Ca2+]i induced by the subsequent addition of sphingosine, OKT3, and PA in the absence of extracellular Ca2+. The present results suggest that at least two kinds of intracellular Ca2+ stores exist in Jurkat cells, both of which are IP3- and PA-sensitive, and that sphingosine mobilizes Ca2+ from both stores in an IP3-independent manner. Furthermore, the IP3- but not the PA-sensitive intracellular Ca2+ store seems to regulate Ca2+ entry induced by sphingosine.
Collapse
Affiliation(s)
- S Sakano
- Department of Pharmacology, School of Medicine, Sapporo Medical University, Japan
| | | | | | | | | | | |
Collapse
|
33
|
Huang WC, Chueh SH. Calcium mobilization from the intracellular mitochondrial and nonmitochondrial stores of the rat cerebellum. Brain Res 1996; 718:151-8. [PMID: 8773778 DOI: 10.1016/0006-8993(96)00108-4] [Citation(s) in RCA: 20] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Two major intracellular Ca2+ stores, the mitochondrial and nonmitochondrial (microsomes) fractions isolated from rat cerebellum exhibited a Ca2+ concentration and ATP-dependent Ca2+ accumulation. The maximal Ca2+ accumulation in mitochondria was higher than in microsomes, but the affinity of the mitochondria for Ca2+ was lower. In this study, Ca2+ accumulation within the mitochondria was energized by ATP hydrolysis. Thus, the protonophore, carbonyl cyanide p-trifluoromethoxyphenylhydrazone, and the F1F0 ATP synthase inhibitor, oligomycin, blocked Ca2+ accumulation and induced the discharge of the entrapped Ca2+ in the mitochondria, whereas the metabolic inhibitor, rotenone, affected neither the Ca2+ accumulation nor discharge. On the other hand, the uniporter inhibitor, ruthenium red, blocked the mitochondrial accumulation of Ca2+, but did not cause the discharge of preloaded Ca2+. In addition, arachidonic acid (AA), sphingosylphosphorylcholine (SPC) and sphingosine (SPH) elicited the dose-dependent release of Ca2+ from microsomal stores. Although the magnitudes of the Ca2+ release induced by AA, SPC or SPH were all dependent on the presence of extravesicular Ca2+ at concentrations ranging from 0.01 to 0.1 microM Ca2+, only the AA- and SPC-evoked Ca2+ releases were insensitive to temperature. The mitochondria were more sensitive than the microsomes to the AA induced release of accumulated Ca2+. Our results indicate the existence of multiple intracellular Ca2+ stores in nerve cells which can be released by various Ca2+ mediators.
Collapse
Affiliation(s)
- W C Huang
- Department of Biochemistry, National Defense Medical Center, Taipei, Taiwan
| | | |
Collapse
|
34
|
Mao C, Kim SH, Almenoff JS, Rudner XL, Kearney DM, Kindman LA. Molecular cloning and characterization of SCaMPER, a sphingolipid Ca2+ release-mediating protein from endoplasmic reticulum. Proc Natl Acad Sci U S A 1996; 93:1993-6. [PMID: 8700873 PMCID: PMC39897 DOI: 10.1073/pnas.93.5.1993] [Citation(s) in RCA: 73] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
Release of Ca2+ stored in endoplasmic reticulum is a ubiquitous mechanism involved in cellular signal transduction, proliferation, and apoptosis. Recently, sphingolipid metabolites have been recognized as mediators of intracellular Ca2+ release, through their action at a previously undescribed intracellular Ca2+ channel. Here we describe the molecular cloning and characterization of a protein that causes the expression of sphingosyl-phosphocholine-mediated Ca2+ release when its complementary RNA is injected into Xenopus oocytes. SCaMPER (for sphingolipid Ca2+ release-mediating protein of endoplasmic reticulum) is an 181 amino acid protein with two putative membrane-spanning domains. SCaMPER is incorporated into microsomes upon expression in SO cells or after translation in vitro. It mediates Ca2+ release at 4 degrees C as well as 22 degrees C, consistent with having ion channel function. The EC50 for Ca2+ release from Xenopus oocytes is 40 microM, similar to sphingosyl-phosphocholine-mediated Ca2+ release from permeabilized mammalian cells. Because Ca2+ release is not blocked by ryanodine or La3+, the activity described here is distinct from the Ca2+ release activity of the ryanodine receptor and the inositol 1,4,5-trisphosphate receptor. The properties of SCaMPER are identical to those of the sphingolipid-gated Ca2+ channel that we have previously described. These findings suggest that SCaMPER is a sphingolipid-gated Ca2+-permeable channel and support its role as a mediator of this pathway for intracellular Ca2+ signal transduction.
Collapse
Affiliation(s)
- C Mao
- Program in Molecular Medicine, Department of Medicine, Duke University Medical Center, Durham, NC 27710, USA
| | | | | | | | | | | |
Collapse
|
35
|
van Koppen C, Meyer zu Heringdorf M, Laser KT, Zhang C, Jakobs KH, Bünemann M, Pott L. Activation of a high affinity Gi protein-coupled plasma membrane receptor by sphingosine-1-phosphate. J Biol Chem 1996; 271:2082-7. [PMID: 8567663 DOI: 10.1074/jbc.271.4.2082] [Citation(s) in RCA: 158] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
Sphingosine-1-phosphate (SPP) has attracted much attention as a possible second messenger controlling cell proliferation and motility and as an intracellular Ca(2+)-releasing agent. Here, we present evidence that SPP activates a G protein-coupled receptor in the plasma membrane of various cells, leading to increase in cytoplasmic Ca2+ concentration ([Ca2+]i), inhibition of adenylyl cyclase, and opening of G protein-regulated potassium channels. In human enbryonic kidney (HEK) cells, SPP potently (EC50, 2 nM) and rapidly increased [Ca2+]i in a pertussis toxin-sensitive manner. Pertussis toxin-sensitive increase in [Ca2+]i was also observed with sphingosylphosphorylcholine (EC50, 460 nM), whereas other sphingolipids, including ceramide-1-phosphate, N-palmitoyl-sphingosine, psychosine, and D-erythro-sphingosine at micromolar concentrations did not or only marginally increased [Ca2+]i. Furthermore, SPP inhibited forskolin-stimulated cAMP accumulation in HEK cells and increased binding of guanosine 5'3-O-(thio) triphosphate to HEK cell membranes. Rapid [Ca2+]i responses were also observed in human transitional bladder carcinoma (J82) cells, monkey COS-1 cells, mouse NIH 3T3 cells, Chinese hamster ovary (CHO-K1) cells, and rat C6 glioma cells, whereas human HL-60 leukemia cells and human erythroleukemia cells failed to respond to SPP. In guinea pig atrial myocytes, SPP activated Gi protein-regulated inwardly rectifying potassium channels. Activation of these channels occurred strictly when SPP was applied at the extracellular face of atrial myocyte plasma membrane as measured in cell-attached and inside-out patch clamp current recordings. We conclude that SPP, in addition to its proposed direct action on intracellular Ca2+ stores, interacts with a high affinity Gi protein-coupled receptor in the plasma membrane of apparently many different cell types.
Collapse
Affiliation(s)
- C van Koppen
- Institut für Pharmakologie, Universität GH Essen, Germany
| | | | | | | | | | | | | |
Collapse
|