1
|
Jong YI, Harmon SK, O'Malley KL. GPCR
Signaling from Intracellular Membranes. GPCRS AS THERAPEUTIC TARGETS 2022:216-298. [DOI: 10.1002/9781119564782.ch8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2025]
|
2
|
Barnes DA, Galloway DA, Hoener MC, Berry MD, Moore CS. TAAR1 Expression in Human Macrophages and Brain Tissue: A Potential Novel Facet of MS Neuroinflammation. Int J Mol Sci 2021; 22:ijms222111576. [PMID: 34769007 PMCID: PMC8584001 DOI: 10.3390/ijms222111576] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 10/24/2021] [Accepted: 10/25/2021] [Indexed: 11/16/2022] Open
Abstract
TAAR1 is a neuroregulator with emerging evidence suggesting a role in immunomodulation. Multiple sclerosis (MS) is an immune-mediated demyelinating disease of the central nervous system. Here, we investigate TAAR1 expression in human primary monocytes, peripherally-derived macrophages, and MS brain tissue. RT-qPCR was used to assess TAAR1 levels in MS monocytes. Using a previously validated anti-human TAAR1 antibody and fluorescence microscopy, TAAR1 protein was visualized in lipopolysaccharide-stimulated or basal human macrophages, as well as macrophage/microglia populations surrounding, bordering, and within a mixed active/inactive MS lesion. In vivo, TAAR1 mRNA expression was significantly lower in MS monocytes compared to age- and sex-matched healthy controls. In vitro, TAAR1 protein showed a predominant nuclear localization in quiescent/control macrophages with a shift to a diffuse intracellular distribution following lipopolysaccharide-induced activation. In brain tissue, TAAR1 protein was predominantly expressed in macrophages/microglia within the border region of mixed active/inactive MS lesions. Considering that TAAR1-mediated anti-inflammatory effects have been previously reported, decreased mRNA in MS patients suggests possible pathophysiologic relevance. A shift in TAAR1 localization following pro-inflammatory activation suggests its function is altered in pro-inflammatory states, while TAAR1-expressing macrophages/microglia bordering an MS lesion supports TAAR1 as a novel pharmacological target in cells directly implicated in MS neuroinflammation.
Collapse
Affiliation(s)
- David A. Barnes
- Department of Biochemistry, Faculty of Science, Memorial University of Newfoundland, 232 Elizabeth Ave, St. John’s, NL A1B 3X9, Canada; (D.A.B.); (M.D.B.)
- Division of Biomedical Sciences, Faculty of Medicine, Memorial University of Newfoundland, 300 Prince Philip Drive, St. John’s, NL A1B 3V6, Canada;
| | - Dylan A. Galloway
- Division of Biomedical Sciences, Faculty of Medicine, Memorial University of Newfoundland, 300 Prince Philip Drive, St. John’s, NL A1B 3V6, Canada;
| | - Marius C. Hoener
- Neuroscience, Ophthalmology and Rare Diseases DTA, pRED, Roche Innovation Center Basel, F. Hoffmann-La Roche, 4070 Basel, Switzerland;
| | - Mark D. Berry
- Department of Biochemistry, Faculty of Science, Memorial University of Newfoundland, 232 Elizabeth Ave, St. John’s, NL A1B 3X9, Canada; (D.A.B.); (M.D.B.)
| | - Craig S. Moore
- Division of Biomedical Sciences, Faculty of Medicine, Memorial University of Newfoundland, 300 Prince Philip Drive, St. John’s, NL A1B 3V6, Canada;
- Correspondence: ; Tel.: +1-709-864-4955
| |
Collapse
|
3
|
Alessio N, Squillaro T, Lettiero I, Galano G, De Rosa R, Peluso G, Galderisi U, Di Bernardo G. Biomolecular Evaluation of Piceatannol's Effects in Counteracting the Senescence of Mesenchymal Stromal Cells: A New Candidate for Senotherapeutics? Int J Mol Sci 2021; 22:ijms222111619. [PMID: 34769049 PMCID: PMC8583715 DOI: 10.3390/ijms222111619] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Revised: 10/22/2021] [Accepted: 10/25/2021] [Indexed: 11/16/2022] Open
Abstract
Several investigations on senescence and its causative role in aging have underscored the importance of developing senotherapeutics, a field focused on killing senescent cells and/or preventing their accumulation within tissues. Using polyphenols in counteracting senescence may facilitate the development of senotherapeutics given their presence in the human diet, their confirmed tolerability and absence of severe side effects, and their role in preventing senescence and inducing the death of senescent cells. Against that background, we evaluated the effect of piceatannol, a natural polyphenol, on the senescence of mesenchymal stromal cells (MSCs), which play a key role in the body's homeostasis. Among our results, piceatannol reduced the number of senescent cells both after genotoxic stress that induced acute senescence and in senescent replicative cultures. Such senotherapeutics activity, moreover, promoted the recovery of cell proliferation and the stemness properties of MSCs. Altogether, our findings demonstrate piceatannol's effectiveness in counteracting senescence by targeting its associated pathways and detecting and affecting P53-dependent and P53-independent senescence. Our study thus suggests that, given piceatannol's various mechanisms to accomplish its pleiotropic activities, it may be able to counteract any senescent phenotypes.
Collapse
Affiliation(s)
- Nicola Alessio
- Department of Experimental Medicine, Biotechnology and Molecular Biology Section, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (N.A.); (T.S.); (I.L.)
| | - Tiziana Squillaro
- Department of Experimental Medicine, Biotechnology and Molecular Biology Section, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (N.A.); (T.S.); (I.L.)
| | - Ida Lettiero
- Department of Experimental Medicine, Biotechnology and Molecular Biology Section, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (N.A.); (T.S.); (I.L.)
| | - Giovanni Galano
- ASL Napoli 1 Centro P.S.I. Napoli Est-Barra, 80147 Naples, Italy; (G.G.); (R.D.R.)
| | - Roberto De Rosa
- ASL Napoli 1 Centro P.S.I. Napoli Est-Barra, 80147 Naples, Italy; (G.G.); (R.D.R.)
| | - Gianfranco Peluso
- Research Institute on Terrestrial Ecosystems, CNR, 80131 Naples, Italy;
| | - Umberto Galderisi
- Department of Experimental Medicine, Biotechnology and Molecular Biology Section, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (N.A.); (T.S.); (I.L.)
- Center for Biotechnology, Sbarro Institute for Cancer Research and Molecular Medicine, Temple University, Philadelphia, PA 19122, USA
- Correspondence: (U.G.); (G.D.B.)
| | - Giovanni Di Bernardo
- Department of Experimental Medicine, Biotechnology and Molecular Biology Section, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (N.A.); (T.S.); (I.L.)
- Center for Biotechnology, Sbarro Institute for Cancer Research and Molecular Medicine, Temple University, Philadelphia, PA 19122, USA
- Correspondence: (U.G.); (G.D.B.)
| |
Collapse
|
4
|
Gonçalves-Monteiro S, Ribeiro-Oliveira R, Vieira-Rocha MS, Vojtek M, Sousa JB, Diniz C. Insights into Nuclear G-Protein-Coupled Receptors as Therapeutic Targets in Non-Communicable Diseases. Pharmaceuticals (Basel) 2021; 14:439. [PMID: 34066915 PMCID: PMC8148550 DOI: 10.3390/ph14050439] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 04/27/2021] [Accepted: 04/30/2021] [Indexed: 12/14/2022] Open
Abstract
G-protein-coupled receptors (GPCRs) comprise a large protein superfamily divided into six classes, rhodopsin-like (A), secretin receptor family (B), metabotropic glutamate (C), fungal mating pheromone receptors (D), cyclic AMP receptors (E) and frizzled (F). Until recently, GPCRs signaling was thought to emanate exclusively from the plasma membrane as a response to extracellular stimuli but several studies have challenged this view demonstrating that GPCRs can be present in intracellular localizations, including in the nuclei. A renewed interest in GPCR receptors' superfamily emerged and intensive research occurred over recent decades, particularly regarding class A GPCRs, but some class B and C have also been explored. Nuclear GPCRs proved to be functional and capable of triggering identical and/or distinct signaling pathways associated with their counterparts on the cell surface bringing new insights into the relevance of nuclear GPCRs and highlighting the nucleus as an autonomous signaling organelle (triggered by GPCRs). Nuclear GPCRs are involved in physiological (namely cell proliferation, transcription, angiogenesis and survival) and disease processes (cancer, cardiovascular diseases, etc.). In this review we summarize emerging evidence on nuclear GPCRs expression/function (with some nuclear GPCRs evidencing atypical/disruptive signaling pathways) in non-communicable disease, thus, bringing nuclear GPCRs as targets to the forefront of debate.
Collapse
Affiliation(s)
- Salomé Gonçalves-Monteiro
- Laboratory of Pharmacology, Department of Drug Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal; (S.G.-M.); (R.R.-O.); (M.S.V.-R.); (M.V.)
- LAQV/REQUIMTE, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
| | - Rita Ribeiro-Oliveira
- Laboratory of Pharmacology, Department of Drug Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal; (S.G.-M.); (R.R.-O.); (M.S.V.-R.); (M.V.)
- LAQV/REQUIMTE, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
| | - Maria Sofia Vieira-Rocha
- Laboratory of Pharmacology, Department of Drug Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal; (S.G.-M.); (R.R.-O.); (M.S.V.-R.); (M.V.)
- LAQV/REQUIMTE, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
| | - Martin Vojtek
- Laboratory of Pharmacology, Department of Drug Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal; (S.G.-M.); (R.R.-O.); (M.S.V.-R.); (M.V.)
- LAQV/REQUIMTE, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
| | - Joana B. Sousa
- Laboratory of Pharmacology, Department of Drug Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal; (S.G.-M.); (R.R.-O.); (M.S.V.-R.); (M.V.)
- LAQV/REQUIMTE, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
| | - Carmen Diniz
- Laboratory of Pharmacology, Department of Drug Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal; (S.G.-M.); (R.R.-O.); (M.S.V.-R.); (M.V.)
- LAQV/REQUIMTE, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
| |
Collapse
|
5
|
Mohammad Nezhady MA, Rivera JC, Chemtob S. Location Bias as Emerging Paradigm in GPCR Biology and Drug Discovery. iScience 2020; 23:101643. [PMID: 33103080 PMCID: PMC7569339 DOI: 10.1016/j.isci.2020.101643] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
GPCRs are the largest receptor family that are involved in virtually all biological processes. Pharmacologically, they are highly druggable targets, as they cover more than 40% of all drugs in the market. Our knowledge of biased signaling provided insight into pharmacology vastly improving drug design to avoid unwanted effects and achieve higher efficacy and selectivity. However, yet another feature of GPCR biology is left largely unexplored, location bias. Recent developments in this field show promising avenues for evolution of new class of pharmaceuticals with greater potential for higher level of precision medicine. Further consideration and understanding of this phenomenon with deep biochemical and molecular insights would pave the road to success. In this review, we critically analyze this perspective and discuss new avenues of investigation.
Collapse
Affiliation(s)
- Mohammad Ali Mohammad Nezhady
- Programmes en Biologie Moléculaire, Faculté de Médecine, Université de Montréal, Montreal, QC, Canada
- Centre de Recherche du CHU Sainte-Justine, Montreal, QC H3T 1C5, Canada
- Corresponding author
| | | | - Sylvain Chemtob
- Programmes en Biologie Moléculaire, Faculté de Médecine, Université de Montréal, Montreal, QC, Canada
- Centre de Recherche du CHU Sainte-Justine, Montreal, QC H3T 1C5, Canada
- Maisonneuve-Rosemont Hospital Research Centre, Montreal, QC, Canada
| |
Collapse
|
6
|
Ribeiro-Oliveira R, Vojtek M, Gonçalves-Monteiro S, Vieira-Rocha MS, Sousa JB, Gonçalves J, Diniz C. Nuclear G-protein-coupled receptors as putative novel pharmacological targets. Drug Discov Today 2019; 24:2192-2201. [DOI: 10.1016/j.drudis.2019.09.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Revised: 08/15/2019] [Accepted: 09/05/2019] [Indexed: 12/14/2022]
|
7
|
Gendron DR, Lecours PB, Lemay AM, Beaulieu MJ, Huppé CA, Lee-Gosselin A, Flamand N, Don AS, Bissonnette É, Blanchet MR, Laplante M, Bourgoin SG, Bossé Y, Marsolais D. A Phosphorylatable Sphingosine Analog Induces Airway Smooth Muscle Cytostasis and Reverses Airway Hyperresponsiveness in Experimental Asthma. Front Pharmacol 2017; 8:78. [PMID: 28270767 PMCID: PMC5318459 DOI: 10.3389/fphar.2017.00078] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2016] [Accepted: 02/07/2017] [Indexed: 11/13/2022] Open
Abstract
In asthma, excessive bronchial narrowing associated with thickening of the airway smooth muscle (ASM) causes respiratory distress. Numerous pharmacological agents prevent experimental airway hyperresponsiveness (AHR) when delivered prophylactically. However, most fail to resolve this feature after disease is instated. Although sphingosine analogs are primarily perceived as immune modulators with the ability to prevent experimental asthma, they also influence processes associated with tissue atrophy, supporting the hypothesis that they could interfere with mechanisms sustaining pre-established AHR. We thus assessed the ability of a sphingosine analog (AAL-R) to reverse AHR in a chronic model of asthma. We dissected the pharmacological mechanism of this class of agents using the non-phosphorylatable chiral isomer AAL-S and the pre-phosphorylated form of AAL-R (AFD-R) in vivo and in human ASM cells. We found that a therapeutic course of AAL-R reversed experimental AHR in the methacholine challenge test, which was not replicated by dexamethasone or the non-phosphorylatable isomer AAL-S. AAL-R efficiently interfered with ASM cell proliferation in vitro, supporting the concept that immunomodulation is not necessary to interfere with cellular mechanisms sustaining AHR. Moreover, the sphingosine-1-phosphate lyase inhibitor SM4 and the sphingosine-1-phosphate receptor antagonist VPC23019 failed to inhibit proliferation, indicating that intracellular accumulation of sphingosine-1-phosphate or interference with cell surface S1P1/S1P3 activation, are not sufficient to induce cytostasis. Potent AAL-R-induced cytostasis specifically related to its ability to induce intracellular AFD-R accumulation. Thus, a sphingosine analog that possesses the ability to be phosphorylated in situ interferes with cellular mechanisms that beget AHR.
Collapse
Affiliation(s)
- David R Gendron
- Centre de Recherche de l'Institut Universitaire de Cardiologie et de Pneumologie de Québec, Québec QC, Canada
| | - Pascale B Lecours
- Centre de Recherche de l'Institut Universitaire de Cardiologie et de Pneumologie de Québec, Québec QC, Canada
| | - Anne-Marie Lemay
- Centre de Recherche de l'Institut Universitaire de Cardiologie et de Pneumologie de Québec, Québec QC, Canada
| | - Marie-Josée Beaulieu
- Centre de Recherche de l'Institut Universitaire de Cardiologie et de Pneumologie de Québec, Québec QC, Canada
| | - Carole-Ann Huppé
- Centre de Recherche de l'Institut Universitaire de Cardiologie et de Pneumologie de Québec, Québec QC, Canada
| | - Audrey Lee-Gosselin
- Centre de Recherche de l'Institut Universitaire de Cardiologie et de Pneumologie de Québec, Québec QC, Canada
| | - Nicolas Flamand
- Centre de Recherche de l'Institut Universitaire de Cardiologie et de Pneumologie de Québec, QuébecQC, Canada; Faculty of Medicine, Université Laval, QuébecQC, Canada
| | - Anthony S Don
- Centenary Institute and NHMRC Clinical Trials Centre, University of Sydney, Camperdown NSW, Australia
| | - Élyse Bissonnette
- Centre de Recherche de l'Institut Universitaire de Cardiologie et de Pneumologie de Québec, QuébecQC, Canada; Faculty of Medicine, Université Laval, QuébecQC, Canada
| | - Marie-Renée Blanchet
- Centre de Recherche de l'Institut Universitaire de Cardiologie et de Pneumologie de Québec, QuébecQC, Canada; Faculty of Medicine, Université Laval, QuébecQC, Canada
| | - Mathieu Laplante
- Centre de Recherche de l'Institut Universitaire de Cardiologie et de Pneumologie de Québec, QuébecQC, Canada; Faculty of Medicine, Université Laval, QuébecQC, Canada
| | - Sylvain G Bourgoin
- Faculty of Medicine, Université Laval, QuébecQC, Canada; Division of Infectious Diseases and Immunology, CHU de Québec Research Center, QuébecQC, Canada
| | - Ynuk Bossé
- Centre de Recherche de l'Institut Universitaire de Cardiologie et de Pneumologie de Québec, QuébecQC, Canada; Faculty of Medicine, Université Laval, QuébecQC, Canada
| | - David Marsolais
- Centre de Recherche de l'Institut Universitaire de Cardiologie et de Pneumologie de Québec, QuébecQC, Canada; Faculty of Medicine, Université Laval, QuébecQC, Canada
| |
Collapse
|
8
|
Sphingosine-1-Phosphate Signaling in Immune Cells and Inflammation: Roles and Therapeutic Potential. Mediators Inflamm 2016; 2016:8606878. [PMID: 26966342 PMCID: PMC4761394 DOI: 10.1155/2016/8606878] [Citation(s) in RCA: 109] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2015] [Accepted: 01/03/2016] [Indexed: 12/26/2022] Open
Abstract
Sphingosine-1-phosphate (S1P) is a bioactive sphingolipid metabolite involved in many critical cell processes. It is produced by the phosphorylation of sphingosine by sphingosine kinases (SphKs) and exported out of cells via transporters such as spinster homolog 2 (Spns2). S1P regulates diverse physiological processes by binding to specific G protein-binding receptors, S1P receptors (S1PRs) 1-5, through a process coined as "inside-out signaling." The S1P concentration gradient between various tissues promotes S1PR1-dependent migration of T cells from secondary lymphoid organs into the lymphatic and blood circulation. S1P suppresses T cell egress from and promotes retention in inflamed peripheral tissues. S1PR1 in T and B cells as well as Spns2 in endothelial cells contributes to lymphocyte trafficking. FTY720 (Fingolimod) is a functional antagonist of S1PRs that induces systemic lymphopenia by suppression of lymphocyte egress from lymphoid organs. In this review, we summarize previous findings and new discoveries about the importance of S1P and S1PR signaling in the recruitment of immune cells and lymphocyte retention in inflamed tissues. We also discuss the role of S1P-S1PR1 axis in inflammatory diseases and wound healing.
Collapse
|
9
|
Dukala DE, Soliven B. S1P1deletion in oligodendroglial lineage cells: Effect on differentiation and myelination. Glia 2015; 64:570-82. [DOI: 10.1002/glia.22949] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2015] [Revised: 10/14/2015] [Accepted: 11/17/2015] [Indexed: 01/21/2023]
Affiliation(s)
- Danuta E. Dukala
- Department of Neurology; the University of Chicago; Chicago Illinois
| | - Betty Soliven
- Department of Neurology; the University of Chicago; Chicago Illinois
| |
Collapse
|
10
|
Chen JJ, Lin DJQ, Liu MSY, Chien EJ. Non-genomic rapid responses via progesterone in human peripheral T cells are not indirectly mimicked by sphingosine 1-phosphate. Steroids 2014; 81:9-12. [PMID: 24269742 DOI: 10.1016/j.steroids.2013.11.011] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
Progesterone is an endogenous immunomodulator that suppresses T cell activation during pregnancy. Progesterone has been shown to induce rapid responses that cause intracellular calcium ([Ca(2+)]i) elevation and acidification followed by inhibition of phytohemagglutinin (PHA)-stimulated proliferation. These rapid responses involve T cell plasma membrane sites, but the mechanisms remain unclear. Three new membrane progesterone receptors (mPRα/mPRβ/mPRγ) have been identified as expressed in T cells. These proteins have been identified as G-protein-coupled receptors. Recently, mPRs have been classified as progestin and adipoQ receptors (PAQRs). Furthermore, they have been suggested to be alkaline ceramidases, possibly involved in mediating sphingolipid signaling. Alkaline ceramidases are capable of converting ceramide to sphingosine, which might then be further phosphorylated sphingosine via sphingosine kinase to sphingosine 1-phosphate (S1P). This pathway could result in progesterone acting indirectly via S1P on membrane sphingosine 1-phosphate receptors (S1PRs) in T cells to induce rapid responses. Therefore, our aim was to investigate whether progesterone rapid responses occur indirectly in T cells via S1P. We found that S1P induces [Ca(2+)]i elevation however there was no change in intracellular pH. This is different from the situation with progesterone: S1P alone does not suppress PHA-stimulated cell proliferation and does not act synergistically with progesterone on the inhibition of PHA-induced cell proliferation. In contrast, S1P at 1μM is able to antagonize the proliferation inhibitory effect of progesterone. Thus the rapid responses that are induced by progesterone in human peripheral T cells probably do not involve indirect signaling via S1P and S1PRs.
Collapse
Affiliation(s)
- Jiann-Jong Chen
- Department of Nursing, Cardinal Tien Junior College of Healthcare and Management, Sindian District, New Taipei City 23143, Taiwan, ROC
| | - David Jia-Qing Lin
- Institute of Physiology, School of Medicine, National Yang-Ming University, Taipei 11221, Taiwan, ROC
| | - Mark Shui-Yu Liu
- Institute of Physiology, School of Medicine, National Yang-Ming University, Taipei 11221, Taiwan, ROC
| | - Eileen Jea Chien
- Institute of Physiology, School of Medicine, National Yang-Ming University, Taipei 11221, Taiwan, ROC.
| |
Collapse
|
11
|
Ohotski J, Rosen H, Bittman R, Pyne S, Pyne NJ. Sphingosine kinase 2 prevents the nuclear translocation of sphingosine 1-phosphate receptor-2 and tyrosine 416 phosphorylated c-Src and increases estrogen receptor negative MDA-MB-231 breast cancer cell growth: The role of sphingosine 1-phosphate receptor-4. Cell Signal 2014; 26:1040-7. [PMID: 24486401 DOI: 10.1016/j.cellsig.2014.01.023] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2013] [Accepted: 01/24/2014] [Indexed: 10/25/2022]
Abstract
We demonstrate that pre-treatment of estrogen receptor negative MDA-MB-231 breast cancer cells containing ectopically expressed HA-tagged sphingosine 1-phosphate receptor-2 (S1P2) with the sphingosine kinase 1/2 inhibitor SKi (2-(p-hydroxyanilino)-4-(p-chlorophenyl)thiazole) or the sphingosine kinase 2 selective inhibitor (R)-FTY720 methyl ether (ROMe) or sphingosine kinase 2 siRNA induced the translocation of HA-tagged S1P2 and Y416 phosphorylated c-Src to the nucleus of these cells. This is associated with reduced growth of HA-tagged S1P2 over-expressing MDA-MB-231 cells. Treatment of HA-S1P2 over-expressing MDA-MB-231 cells with the sphingosine 1-phosphate receptor-4 (S1P4) antagonist CYM50367 or with S1P4 siRNA also promoted nuclear translocation of HA-tagged S1P2. These findings identify for the first time a signaling pathway in which sphingosine 1-phosphate formed by sphingosine kinase 2 binds to S1P4 to prevent nuclear translocation of S1P2 and thereby promote the growth of estrogen receptor negative breast cancer cells.
Collapse
Affiliation(s)
- Jan Ohotski
- Cell Biology Group, Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow G4 0RE, United Kingdom
| | - Hugh Rosen
- The Scripps Research Institute, Department of Chemistry, 10550 N. Torrey Pines Rd., La Jolla, CA 92037, United States
| | - Robert Bittman
- Department of Chemistry and Biochemistry, Queens College, The City University of New York, Flushing, NY 11367-1597, United States
| | - Susan Pyne
- Cell Biology Group, Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow G4 0RE, United Kingdom
| | - Nigel J Pyne
- Cell Biology Group, Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow G4 0RE, United Kingdom.
| |
Collapse
|
12
|
Yin Z, Fan L, Wei L, Gao H, Zhang R, Tao L, Cao F, Wang H. FTY720 protects cardiac microvessels of diabetes: a critical role of S1P1/3 in diabetic heart disease. PLoS One 2012; 7:e42900. [PMID: 22916176 PMCID: PMC3419247 DOI: 10.1371/journal.pone.0042900] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2012] [Accepted: 07/13/2012] [Indexed: 01/07/2023] Open
Abstract
Background: Diabetes is associated with an increased risk of cardiac microvascular disease. The mechanisms by which this damage occurs are unknown. However, research suggests that signaling through the sphingosine-1-phosphates receptor 1 and 3 (S1P1/3) by FTY720, a sphiongolipid drug that is structually similar to SIP, may play a role in the treatment on cardiac microvascular dysfunction in diabetes. We hypothesized that FTY720 might exert the cardioprotective effects of S1P1 and S1P3 viaprotein kinase C-beta (PKCβ II) signaling pathway. Methodology/Principal Findings: Transthoracic echocardiography was performed to detect the change of cardiac function. Scanning and transmission electron microscope with lanthanum tracer were used to determine microvascular ultrastructure and permeability in vivo. Apoptosis was detected by TUNEL and CD31 dual labeling in paraffin-embedded sections. Laser capture miscrodissection was used to assess cardiac micovascular endothelial cells (CMECs) in vivo. RT-PCR and Western blot analysis were used to determine the mRNA levels and protein expression of S1P1, S1P3, and PKCβ II. In the diabetic rats vs. controls, cardiac capillaries showed significantly higher density; CD31 positive endothelial cells were significantly reduced; the apoptosis index of cardiac endothlial cells was significantly higher. And FTY720 could increase the expressional level of S1P1 and boost S1P3 trasnslocation from membrane to nuclear, then ameliorate cardiac microvascular barrier impairment and pathologic angiogenesis induced by diabetes. In addition, overexpression of PKCβ II significantly decreased the protective effect of FTY720. Conclusions: Our study represents that the deregulation of S1P1 and S1P3 is an important signalresponsible for cardiac microvascular dysfunction in diabetes. FTY720 might be competent to serve as a potential therapeutic approach for diabetic heart disease through ameliorating cardiac microvascular barrier impairment and pathologic angiogenesis, which might be partly dependent on PKCβII-mediated signaling pathway.
Collapse
Affiliation(s)
- Zhiyong Yin
- Department of Cardiology, Xijing Hospital, The Fourth Military Medical University, Xi’an City, Shaanxi Province, China
| | - Linni Fan
- Department of Pathology, Xijing Hospital, The Fourth Military Medical University, Xi’an City, Shaanxi Province, China
| | - Liping Wei
- Department of Cardiology, Tianjin Union Medicine Center, Tianjin, China
| | - Haokao Gao
- Department of Cardiology, Xijing Hospital, The Fourth Military Medical University, Xi’an City, Shaanxi Province, China
| | - Rongqing Zhang
- Department of Cardiology, Xijing Hospital, The Fourth Military Medical University, Xi’an City, Shaanxi Province, China
| | - Ling Tao
- Department of Cardiology, Xijing Hospital, The Fourth Military Medical University, Xi’an City, Shaanxi Province, China
| | - Feng Cao
- Department of Cardiology, Xijing Hospital, The Fourth Military Medical University, Xi’an City, Shaanxi Province, China
| | - Haichang Wang
- Department of Cardiology, Xijing Hospital, The Fourth Military Medical University, Xi’an City, Shaanxi Province, China
- * E-mail:
| |
Collapse
|
13
|
Ohotski J, Edwards J, Elsberger B, Watson C, Orange C, Mallon E, Pyne S, Pyne NJ. Identification of novel functional and spatial associations between sphingosine kinase 1, sphingosine 1-phosphate receptors and other signaling proteins that affect prognostic outcome in estrogen receptor-positive breast cancer. Int J Cancer 2012; 132:605-16. [DOI: 10.1002/ijc.27692] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2012] [Accepted: 06/11/2012] [Indexed: 12/14/2022]
|
14
|
Ishimaru N, Yamada A, Nitta T, Arakaki R, Lipp M, Takahama Y, Hayashi Y. CCR7 with S1P1 signaling through AP-1 for migration of Foxp3+ regulatory T-cells controls autoimmune exocrinopathy. THE AMERICAN JOURNAL OF PATHOLOGY 2011; 180:199-208. [PMID: 22067914 DOI: 10.1016/j.ajpath.2011.09.027] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/01/2011] [Revised: 08/31/2011] [Accepted: 09/13/2011] [Indexed: 12/16/2022]
Abstract
Forkhead box p3-positive (Foxp3(+)) regulatory T cells (T(reg) cells) participate in maintaining peripheral immune tolerance and suppressing autoimmunity. We recently reported that in situ patrolling by C-C-chemokine receptor 7 (CCR7)(+) T(reg) cells in target organs is essential for controlling autoimmune lesions in Sjögren's syndrome. In the present study, the molecular mechanism underlying CCR7-mediated T(reg) cell migration was investigated in a mouse model. The impaired migratory response of Ccr7(-/-) T(reg) cells to sphingosine 1-phosphate (S1P) occurred because of defective association of S1P receptor 1 (S1P(1)) with a G coupled-protein. In addition, T-cell receptor (TCR)- and S1P(1)-mediated Ras-related C3 botulinum toxin substrate 1 (Rac-1), extracellular signal-related kinase (ERK), and c-Jun phosphorylation required for activator protein 1 (AP-1) transcriptional activity were significantly impaired in Ccr7(-/-) T(reg) cells. Surprisingly, the abnormal nuclear localization of Foxp3 was detected after abrogation of the c-Jun and Foxp3 interaction in the nucleus of Ccr7(-/-) T(reg) cells. These results indicate that CCR7 essentially controls the migratory function of T(reg) cells through S1P(1)-mediated AP-1 signaling, which is regulated through its interaction with Foxp3 in the nucleus.
Collapse
Affiliation(s)
- Naozumi Ishimaru
- Department of Oral Molecular Pathology, Institute of Health Biosciences, The University of Tokushima Graduate School, Tokushima, Japan.
| | | | | | | | | | | | | |
Collapse
|
15
|
Swan DJ, Kirby JA, Ali S. Vascular biology: the role of sphingosine 1-phosphate in both the resting state and inflammation. J Cell Mol Med 2011; 14:2211-22. [PMID: 20716131 PMCID: PMC3822560 DOI: 10.1111/j.1582-4934.2010.01136.x] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
The vascular and immune systems of mammals are closely intertwined: the individual components of the immune system must move between various body compartments to perform their function effectively. Sphingosine 1-phosphate (S1P), a bioactive lipid mediator, exerts effects on the two organ systems and influences the interaction between them. In the resting state, the vascular S1P gradient contributes to control of lymphocyte recirculation through the blood, lymphoid tissue and lymphatic vasculature. The high level of S1P in blood helps maintain endothelial barrier integrity. During the inflammatory process, both the level of S1P in different immune compartments and S1P receptor expression on lymphocytes and endothelial cells are modified, resulting in functionally important changes in endothelial cell and lymphocyte behaviour. These include transient arrest of lymphocytes in secondary lymphoid tissue, crucial for generation of adaptive immunity, and subsequent promotion of lymphocyte recruitment to sites of inflammation. This review begins with an outline of the basic biochemistry of S1P. S1P receptor signalling is then discussed, followed by an exploration of the roles of S1P in the vascular and immune systems, with particular focus on the interface between them. The latter part concerns crosstalk between S1P and other signalling pathways, and concludes with a look at therapies targeting the S1P-S1P receptor axis.
Collapse
Affiliation(s)
- David J Swan
- Applied Immunobiology and Transplantation Research Group, Institute of Cellular Medicine, Newcastle University, Newcastle-upon-Tyne, UK
| | | | | |
Collapse
|
16
|
Magnusson C, Liu J, Ehrnström R, Manjer J, Jirström K, Andersson T, Sjölander A. Cysteinyl leukotriene receptor expression pattern affects migration of breast cancer cells and survival of breast cancer patients. Int J Cancer 2010; 129:9-22. [PMID: 20824707 DOI: 10.1002/ijc.25648] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2010] [Accepted: 08/10/2010] [Indexed: 12/16/2022]
Abstract
The fact that breast cancer patients with local or distal dissemination exhibit decreased survival, promotes a search for novel mechanisms to suppress such tumor progression. Here, we have determined the expression of proinflammatory cysteinyl leukotriene receptors (CysLTRs) in breast tumor tissue and their signaling effect on breast cancer cell functions related to tumor progression. Patients with breast tumors characterized by high CysLT(1)R and low CysLT(2)R expression levels exhibited increased risk of cancer-induced death in univariate analysis for both the total patient group (hazard ratio [HR] = 2.88, 95% confidence interval [CI] = 1.11-7.41), as well as patients with large (>20 mm) tumors (HR = 5.08, 95% CI = 1.39-18.5). Multivariate analysis revealed that patients with large tumors exhibiting high CysLT(1)R and low CysLT(2)R expression levels had a significantly reduced survival, also when adjusted for established prognostic parameters (HR = 7.51, 95% CI = 1.83-30.8). In patients with large (>20 mm) tumors, elevated CysLT(2)R expression predicted an improved 5-year survival (log-rank test p = 0.04). Surprisingly, for longer time periods, this prognostic value was lost. This disappearance coincided with the termination of hormonal treatment. Tamoxifen preserved and even induced transcription of CysLT(2)R, but not CysLT(1)R, in estrogene receptor-positive MCF-7 breast cancer cells. This elevated CysLT(2)R expression decreased, even below the level of untreated cells, when tamoxifen was withdrawn. CysLT(2)R signaling reduced MCF-7 cell migration, but had no effect on either proliferation or apoptosis. Our data indicate that low CysLT(1)R together with high CysLT(2)R expression levels might be useful parameters in prognostication and treatment stratification of breast cancer patients.
Collapse
Affiliation(s)
- Cecilia Magnusson
- Division of Cell and Experimental Pathology, Department of Laboratory Medicine, Lund University, SE-205 02 Malmö, Sweden
| | | | | | | | | | | | | |
Collapse
|
17
|
Abstract
Sphingolipids comprise a complex family of naturally occurring molecules that are enriched in lipid rafts and contribute to their unique biochemical properties. Membrane sphingolipids also serve as a reservoir for bioactive metabolites including sphingosine, ceramide, sphingosine-1-phosphate and ceramide-1-phosphate. Among these, sphingosine-1-phosphate has emerged as a central regulator of mammalian biology. Sphingosine-1-phosphate is essential for mammalian brain and cardiac development and for maturation of the systemic circulatory system and lymphatics. In addition, sphingosine-1-phosphate contributes to trafficking and effector functions of lymphocytes and other hematopoietic cells and protects against various forms of tissue injury. However, sphingosine-1-phosphate is also an oncogenic lipid that promotes tumor growth and progression. Recent preclinical and clinical investigations using pharmacological agents that target sphingosine-1-phosphate, its receptors and the enzymes required for its biosynthesis and degradation demonstrate the promise and potential risks of modulating sphingosine-1-phosphate signaling in treatment strategies for autoimmunity, cancer, cardiovascular disease and other pathological conditions.
Collapse
|
18
|
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
|
19
|
Verzijl D, Peters SLM, Alewijnse AE. Sphingosine-1-phosphate receptors: zooming in on ligand-induced intracellular trafficking and its functional implications. Mol Cells 2010; 29:99-104. [PMID: 20127285 DOI: 10.1007/s10059-010-0041-z] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2009] [Accepted: 12/27/2009] [Indexed: 01/10/2023] Open
Abstract
Regulatory processes including receptor phosphorylation and intracellular trafficking, also referred to as receptor internalization, are important processes to terminate G protein-coupled receptor (GPCR) signaling. Compelling evidence now indicates that internalization of a receptor is not necessarily the endpoint of signaling, but can also be the beginning of the activation of intracellular signaling pathways. Sphingosine-1-phosphate (S1P) receptors, which are activated by the endogenous phospholipid S1P, belong to the family of GPCRs. Interestingly, there is evidence indicating differential intracellular trafficking of one of the S1P receptor subtypes, the S1P1 receptor, upon agonist activation by either S1P or the synthetic agonist FTY720-P. Moreover, the differential effect of FTY720-P on S1P1 receptor regulation has been suggested to be the mechanism of action of this drug, which is now in Phase III clinical trials for the treatment of multiple sclerosis. It is thus of importance to get a good insight into the regulation of S1P receptors. This review therefore gives a detailed overview about the current state of knowledge on S1P receptor internalization and its functional implications, including some data on nuclear signaling of S1P receptors.
Collapse
Affiliation(s)
- Dennis Verzijl
- Department Pharmacology and Pharmacotherapy, Academic Medical Center, Amsterdam, the Netherlands
| | | | | |
Collapse
|
20
|
Sensken SC, Gräler MH. Down-regulation of S1P1 receptor surface expression by protein kinase C inhibition. J Biol Chem 2010; 285:6298-307. [PMID: 20032465 PMCID: PMC2825425 DOI: 10.1074/jbc.m109.049692] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2009] [Revised: 12/18/2009] [Indexed: 11/06/2022] Open
Abstract
The sphingosine 1-phosphate receptor type 1 (S1P(1)) is important for the maintenance of lymphocyte circulation. S1P(1) receptor surface expression on lymphocytes is critical for their egress from thymus and lymph nodes. Premature activation-induced internalization of the S1P(1) receptor in lymphoid organs, mediated either by pharmacological agonists or by inhibition of the S1P degrading enzyme S1P-lyase, blocks lymphocyte egress and induces lymphopenia in blood and lymph. Regulation of S1P(1) receptor surface expression is therefore a promising way to control adaptive immunity. Hence, we analyzed potential cellular targets for their ability to alter S1P(1) receptor surface expression without stimulation. The initial observation that preincubation of mouse splenocytes with its natural analog sphingosine was sufficient to block Transwell chemotaxis to S1P directed subsequent investigations to the underlying mechanism. Sphingosine is known to inhibit protein kinase C (PKC), and PKC inhibition with nanomolar concentrations of staurosporine, calphostin C, and GF109203X down-regulated surface expression of S1P(1) but not S1P(4) in transfected rat hepatoma HTC(4) cells. The PKC activator phorbol 12-myristate 13-acetate partially rescued FTY720-induced down-regulation of the S1P(1) receptor, linking PKC activation with S1P(1) receptor surface expression. FTY720, but not FTY720 phosphate, efficiently inhibited PKC. Cell-based efficacy was obvious with 10 nm FTY720, and in vivo treatment of mice with 0.3-3 mg/kg/day FTY720 showed increasing concentration-dependent effectiveness. PKC inhibition therefore may contribute to lymphopenia by down-regulating S1P(1) receptor cell surface expression independently from its activation.
Collapse
Affiliation(s)
| | - Markus H. Gräler
- From the Institute for Immunology, Hannover Medical School, 30625 Hanover, Germany
| |
Collapse
|
21
|
Gillies L, Lee SC, Long JS, Ktistakis N, Pyne NJ, Pyne S. The sphingosine 1-phosphate receptor 5 and sphingosine kinases 1 and 2 are localised in centrosomes: possible role in regulating cell division. Cell Signal 2009; 21:675-84. [PMID: 19211033 DOI: 10.1016/j.cellsig.2009.01.023] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2008] [Revised: 01/03/2009] [Accepted: 01/04/2009] [Indexed: 11/16/2022]
Abstract
We show here that the endogenous sphingosine 1-phosphate 5 receptor (S1P(5), a G protein coupled receptor (GPCR) whose natural ligand is sphingosine 1-phosphate (S1P)) and sphingosine kinases 1 and 2 (SK1 and SK2), which catalyse formation of S1P, are co-localised in the centrosome of mammalian cells, where they may participate in regulating mitosis. The centrosome is a site for active GTP-GDP cycling involving the G-protein, G(i) and tubulin, which are required for spindle pole organization and force generation during cell division. Therefore, the presence of S1P(5) (which normally functions as a plasma membrane guanine nucleotide exchange factor, GEF) and sphingosine kinases in the centrosome might suggest that S1P(5) may function as a ligand activated GEF in regulating G-protein-dependent spindle formation and mitosis. The addition of S1P to cells inhibits trafficking of S1P(5) to the centrosome, suggesting a dynamic shuttling endocytic mechanism controlled by ligand occupancy of cell surface receptor. We therefore propose that the centrosomal S1P(5) receptor might function as an intracellular target of S1P linked to regulation of mitosis.
Collapse
Affiliation(s)
- Laura Gillies
- Cell Biology Group, SIPBS, University of Strathclyde, 27 Taylor St, Glasgow, G4 0NR, UK
| | | | | | | | | | | |
Collapse
|
22
|
Vogel P, Donoviel MS, Read R, Hansen GM, Hazlewood J, Anderson SJ, Sun W, Swaffield J, Oravecz T. Incomplete inhibition of sphingosine 1-phosphate lyase modulates immune system function yet prevents early lethality and non-lymphoid lesions. PLoS One 2009; 4:e4112. [PMID: 19119317 PMCID: PMC2606024 DOI: 10.1371/journal.pone.0004112] [Citation(s) in RCA: 133] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2008] [Accepted: 12/04/2008] [Indexed: 11/18/2022] Open
Abstract
Background S1PL is an aldehyde-lyase that irreversibly cleaves sphingosine 1-phosphate (S1P) in the terminal step of sphingolipid catabolism. Because S1P modulates a wide range of physiological processes, its concentration must be tightly regulated within both intracellular and extracellular environments. Methodology In order to better understand the function of S1PL in this regulatory pathway, we assessed the in vivo effects of different levels of S1PL activity using knockout (KO) and humanized mouse models. Principal Findings Our analysis showed that all S1PL-deficient genetic models in this study displayed lymphopenia, with sequestration of mature T cells in the thymus and lymph nodes. In addition to the lymphoid phenotypes, S1PL KO mice (S1PL−/−) also developed myeloid cell hyperplasia and significant lesions in the lung, heart, urinary tract, and bone, and had a markedly reduced life span. The humanized knock-in mice harboring one allele (S1PLH/−) or two alleles (S1PLH/H) of human S1PL expressed less than 10 and 20% of normal S1PL activity, respectively. This partial restoration of S1PL activity was sufficient to fully protect both humanized mouse lines from the lethal non-lymphoid lesions that developed in S1PL−/− mice, but failed to restore normal T-cell development and trafficking. Detailed analysis of T-cell compartments indicated that complete absence of S1PL affected both maturation/development and egress of mature T cells from the thymus, whereas low level S1PL activity affected T-cell egress more than differentiation. Significance These findings demonstrate that lymphocyte trafficking is particularly sensitive to variations in S1PL activity and suggest that there is a window in which partial inhibition of S1PL could produce therapeutic levels of immunosuppression without causing clinically significant S1P-related lesions in non-lymphoid target organs.
Collapse
Affiliation(s)
- Peter Vogel
- Lexicon Pharmaceuticals Inc, The Woodlands, TX, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Favre N, Camps M, Arod C, Chabert C, Rommel C, Pasquali C. Chemokine receptor CCR2 undergoes transportin1-dependent nuclear translocation. Proteomics 2008; 8:4560-76. [PMID: 18846510 DOI: 10.1002/pmic.200800211] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Chemokines (CCs) are small chemoattractant cytokines involved in a wide variety of biological and pathological processes. Released by cells in the milieu, and extracellular matrix and activating signalling cascades upon binding to specific G protein-coupled receptors (GPCRs), they trigger many cellular events. In various pathologies, CCs are directly responsible for excessive recruitment of leukocytes to inflammatory sites and recent studies using chemokine receptor (CCR) antagonists permitted these molecules to reach the market for medical use. While interaction of CCs with their receptors has been extensively documented, downstream GPCR signalling cascades triggered by CC are less well understood. Given the pivotal role of chemokine receptor 2 (CCR2) in monocyte recruitment, activation and differentiation and its implication in several autoimmune-inflammatory pathologies, we searched for potential new CCR2-interacting proteins by engineering a modified CCR2 that we used as bait. Herein, we show the direct interaction of CCR2 with transportin1 (TRN1), which we demonstrate is followed by CCR2 receptor internalization. Further characterization of this novel interaction revealed that TRN1-binding to CCR2 increased upon time in agonist treated cells and promotes its nuclear translocation in a TRN1-dependent manner. Finally, we provide evidence that following translocation, the receptor localizes at the outer edge of the nuclear envelope where it is finally released from TRN1.
Collapse
|
24
|
Padmanabhan S, Myers AG, Prasad BM. Constitutively active GPR6 is located in the intracellular compartments. FEBS Lett 2008; 583:107-12. [DOI: 10.1016/j.febslet.2008.11.033] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2008] [Accepted: 11/18/2008] [Indexed: 10/21/2022]
|
25
|
Estrada R, Wang L, Jala VR, Lee JF, Lin CY, Gray RD, Haribabu B, Lee MJ. Ligand-induced nuclear translocation of S1P(1) receptors mediates Cyr61 and CTGF transcription in endothelial cells. Histochem Cell Biol 2008; 131:239-49. [PMID: 18936953 DOI: 10.1007/s00418-008-0521-9] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/24/2008] [Indexed: 01/26/2023]
Abstract
Sphingosine-1-phosphate (S1P) receptor subtype 1 (S1P(1)), a G-protein coupled receptor (GPCR), regulates many biological activities of endothelial cells (ECs). In this report, we show that S1P(1) receptors are present in the nuclei of ECs by using various biochemical and microscopic techniques such as cellular fractionation, immunogold labeling, and confocal microscopic analysis. Live cell imaging showed that plasma membrane S1P(1) receptors are rapidly internalized and subsequently translocated to nuclear compartment upon S1P stimulation. Utilizing membrane biotinylation technique further supports the notion that nuclear S1P(1) receptors were internalized from plasma membrane S1P(1) after ligand treatment. Moreover, nuclear S1P(1) is able to regulate the transcription of Cyr61 and CTGF, two growth factors functionally important in the regulation of vasculature. Collectively, these data suggest a novel S1P-S1P(1) signaling axis present in the nuclear compartment of endothelial cells, which may regulate biological responses of endothelium.
Collapse
Affiliation(s)
- Rosendo Estrada
- Department of Microbiology and Immunology, Gheens Center on Aging, University of Louisville Health Sciences Center, Louisville, KY 40202, USA
| | | | | | | | | | | | | | | |
Collapse
|
26
|
Chiyo M, Iwata T, Webb TJ, Vasko MR, Thompson EL, Heidler KM, Cummings OW, Yoshida S, Fujisawa T, Brand DD, Wilkes DS. Silencing S1P1 receptors regulates collagen-V reactive lymphocyte-mediated immunobiology in the transplanted lung. Am J Transplant 2008; 8:537-46. [PMID: 18294150 DOI: 10.1111/j.1600-6143.2007.02116.x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Type V collagen (col[V])-reactive lymphocytes contribute to lung transplant rejection, but the mechanisms for emigration into the graft are unknown. Sphingosine-1-phosphate-1 receptors (S1P(1R)) are believed to be required for lymphocyte emigration in other studies, but their role in col(V)-reactive lymphocyte rejection responses is not known. Utilizing small interfering RNA (siRNA) to reduce S1P(1R) expression on col(V)-reactive lymphocytes, we examined the role of S1P(1R) in the rejection response. Quantitative polymerase chain reaction (PCR) revealed strong expression of S1P(1R) messenger RNA (mRNA)on col(V)-reactive lymphocytes isolated from immunized rats. S1P(1R)-specific siRNA (S1P(1R) siRNA) reduced expression of S1P(1R) mRNA and protein, whereas scramble siRNA (SC siRNA) had no effect. Adoptive transfer of lymphocytes treated with S1P(1R) siRNA to rat Wistar Kyoto (WKY) lung isograft recipients resulted in retention of cells within the liver with fewer cells in mediastinal lymph nodes when compared to cells exposed to SC siRNA. S1P(1R)-deficient cells proliferated in response to alloantigens, but not in response to col(V), and produced less interferon (IFN)-gamma in response to col(V) compared to controls. Downregulating S1P(1R) did not affect production of interleukin (IL)-10and tumor necrosis factor (TNF)-alpha, or expression of adhesion molecules critical for migration, but prevented rejection pathology and lowered local levels of IFN-gamma post adoptive transfer. These data demonstrate novel roles of S1P(1R,) which include regulating emigration and modulating lymphocyte activation.
Collapse
Affiliation(s)
- M Chiyo
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Activated Protein C Decreases Tumor Necrosis Factor–Related Apoptosis-Inducing Ligand by an EPCR- Independent Mechanism Involving Egr-1/Erk-1/2 Activation. Arterioscler Thromb Vasc Biol 2007; 27:2634-41. [DOI: 10.1161/atvbaha.107.153734] [Citation(s) in RCA: 51] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Background—
APC is an antithrombotic and antiinflammatory serine protease that plays an important role in vascular function. We report that APC can suppress the proapoptotic mediator TRAIL in human umbilical vein endothelial cells, and we have investigated the signaling mechanism.
Methods and Results—
APC inhibited endothelial TRAIL expression and secretion and its induction by cell activation. To explore the mechanism, we examined factors associated with TRAIL regulation and demonstrated that APC increased the level of EGR-1, a transcriptional factor known to suppress the TRAIL promoter. APC also induced a significant increase in phosphorylation of ERK-1/2, required to activate EGR-1 expression. Activation of ERK-1/2 was dependent on the protease activated receptor-1 (PAR-1), but independent of the endothelial protein C receptor (EPCR). Using siRNA, we found that the effect of APC on the EGR-1/ERK signaling required for TRAIL inhibition was dependent on the S1P1 receptor and S1P1 kinase.
Conclusions—
Our data suggest that APC may provide cytoprotective activity by activating the ERK pathway, which upregulates EGR-1 thereby suppressing the expression of TRAIL. Moreover, we provide evidence that APC can induce a cell signaling response through a PAR-1/S1P1-dependent but EPCR-independent mechanism.
Collapse
|
28
|
Liao JJ, Huang MC, Goetzl EJ. Cutting edge: Alternative signaling of Th17 cell development by sphingosine 1-phosphate. THE JOURNAL OF IMMUNOLOGY 2007; 178:5425-8. [PMID: 17442922 DOI: 10.4049/jimmunol.178.9.5425] [Citation(s) in RCA: 98] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Sphingosine 1-phosphate (S1P) in blood and lymph controls T cell traffic and proliferation through type 1 S1P receptor (S1P(1)) signals, but suppression of IFN-gamma generation has been the only consistently observed effect on T cell cytokines. The fact that S1P enhances the development of Th17 cells from Ag-challenged transgenic S1P(1)-overexpressing CD4 T cells suggested that the S1P-S1P(1) axis may promote the expansion of Th17 cells in wild-type mice. In a model of Th17 cell development from CD4 T cells stimulated by anti-CD3 plus anti-CD28 Abs and a mixture of TGF-beta1, IL-1, and IL-6, S1P enhanced their number and IL-17-generating activity the same as IL-23. As for IL-23 enhancement of Th17 cell development, that by S1P was prevented by IL-4 plus IFN-gamma and by IL-27. The prevention of S1P augmentation of Th17 cell development by the S1P receptor agonist and down-regulator FTY720 implies that FTY720 immunosuppression is attributable partially to inhibition of Th17-mediated inflammation.
Collapse
Affiliation(s)
- Jia-Jun Liao
- Department of Medicine and Department of Microbiology-Immunology, University of California, San Francisco, CA 94143, USA
| | | | | |
Collapse
|
29
|
Brinkmann V. Sphingosine 1-phosphate receptors in health and disease: mechanistic insights from gene deletion studies and reverse pharmacology. Pharmacol Ther 2007; 115:84-105. [PMID: 17561264 DOI: 10.1016/j.pharmthera.2007.04.006] [Citation(s) in RCA: 437] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2007] [Accepted: 04/20/2007] [Indexed: 12/21/2022]
Abstract
Sphingosine 1-phosphate (S1P) is a bioactive sphingolipid that is critically involved in the embryonic development of the cardiovascular and central nervous systems. In the adult, S1P can produce cytoskeletal re-arrangements in many cell types to regulate immune cell trafficking, vascular homeostasis and cell communication in the central nervous system. S1P is contained in body fluids and tissues at different concentrations, and excessive production of the pleiotropic mediator at inflammatory sites may participate in various pathological conditions. Gene deletion studies and reverse pharmacology (techniques aiming to identify both ligands and function of receptors) provided evidence that many effects of S1P are mediated via five G-protein-coupled S1P receptor subtypes, and novel therapeutic strategies based on interaction with these receptors are being initiated. The prototype S1P receptor modulator, FTY720 (fingolimod), targets four of the five S1P receptor subtypes and may act at several levels to modulate lymphocyte trafficking via lymphocytic and endothelial S1P1 and, perhaps, other inflammatory processes through additional S1P receptor subtypes. A recently completed Phase II clinical trial suggested that the drug may provide an effective treatment of relapsing-remitting multiple sclerosis. FTY720 is currently being evaluated in larger-scale, longer-term, Phase III studies. This review provides an overview on S1P activities and S1P receptor function in health and disease, and summarizes the clinical experience with FTY720 in transplantation and multiple sclerosis.
Collapse
Affiliation(s)
- Volker Brinkmann
- Autoimmunity and Transplantation, Novartis Institutes for BioMedical Research, Lichtstrasse 35, CH-4002 Basel, Switzerland.
| |
Collapse
|
30
|
Goetzl EJ, Wang W, McGiffert C, Liao JJ, Huang MC. Sphingosine 1-phosphate as an intracellular messenger and extracellular mediator in immunity. Acta Paediatr 2007; 96:49-52. [PMID: 17391442 DOI: 10.1111/j.1651-2227.2007.00208.x] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
UNLABELLED The omnific mediator system composed of sphingosine 1-phosphate (S1P) and its five G-protein-coupled receptors, designated S1P(1)-S1P(5), affects diverse cellular functions in the nervous, endocrine, cardiovascular and immune systems. The many activities of the S1P-S1P(1) axis, which predominates in the cardiovascular and immune systems, have previously been classified according to their relationship with the distinct functional roles of each type of cell or according to their most frequently used signalling pathways. In the immune system, cell surface S1P(1) receptors transduce the rapid, transient effects of extracellular S1P on T- and B-lymphocyte trafficking in the lymphoid system, lymphocyte migration in non-immune tissues and cytokine generation. After immune stimulation of T- and B-lymphocytes, S1P(1) receptors translocate from the cell surface to endosomal and nuclear compartments. The present hypothesis is that nuclear S1P(1) receptors represent distinct signalling complexes that, through a series of transcriptional events, transduce the sustained effects of intracellular S1P on survival and proliferation of T-lymphocytes. It is postulated that similar types of sustained signalling from nuclear S1P receptors in other types of cells affect proliferation, survival and specific effector functional activities. Effective pharmacological approaches to intracellular, as well as cell surface, S1P-S1P receptor axes will thus require the bioaccessibility of agonists and antagonists to the nuclear domain of relevant target cells. CONCLUSION Most investigations of the effects of the S1P-S1P(1) axis in immunity have focused primarily on rapid, transient alterations in lymphocyte migration and trafficking, and on mast cell migration and secretion of chemical mediators. The discovery of functional S1P(1)-G protein signalling complexes in the nuclear membranes of activated lymphocytes, that are coupled to the transduction of prolonged inhibition of proliferative responses by intracellular S1P, adds a new dimension to the role of the S1P-S1P(1) axis in immunity. Recruitment of this novel, potentially immunosuppressive, function of S1P(1) may be beneficial in some autoimmune diseases and will require application of cell membrane-permeant S1P(1)-specific drugs.
Collapse
Affiliation(s)
- Edward J Goetzl
- Department of Medicine, University of California, San Francisco, CA 94143-0711, USA.
| | | | | | | | | |
Collapse
|
31
|
Chapter 16 Sphingosine 1-Phosphate Type 1 Receptor Modulators: Recent Advances and Therapeutic Potential. ANNUAL REPORTS IN MEDICINAL CHEMISTRY 2007. [DOI: 10.1016/s0065-7743(07)42016-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2023]
|