Sphingosine 1-Phosphate Receptor Modulators and Drug Discovery

Total Page:16

File Type:pdf, Size:1020Kb

Sphingosine 1-Phosphate Receptor Modulators and Drug Discovery Invited Review Biomol Ther 25(1), 80-90 (2017) Sphingosine 1-Phosphate Receptor Modulators and Drug Discovery Soo-Jin Park and Dong-Soon Im* Molecular Inflammation Research Center for Aging Intervention (MRCA) and College of Pharmacy, Pusan National University, Busan 46241, Republic of Korea Abstract Initial discovery on sphingosine 1-phosphate (S1P) as an intracellular second messenger was faced unexpectedly with roles of S1P as a first messenger, which subsequently resulted in cloning of its G protein-coupled receptors, S1P1-5. The molecular iden- tification of S1P receptors opened up a new avenue for pathophysiological research on this lipid mediator. Cellular and molecular in vitro studies and in vivo studies on gene deficient mice have elucidated cellular signaling pathways and the pathophysiological meanings of S1P receptors. Another unexpected finding that fingolimod (FTY720) modulates S1P receptors accelerated drug discovery in this field. Fingolimod was approved as a first-in-class, orally active drug for relapsing multiple sclerosis in 2010, and its applications in other disease conditions are currently under clinical trials. In addition, more selective S1P receptor modulators with better pharmacokinetic profiles and fewer side effects are under development. Some of them are being clinically tested in the contexts of multiple sclerosis and other autoimmune and inflammatory disorders, such as, psoriasis, Crohn’s disease, ulcerative colitis, polymyositis, dermatomyositis, liver failure, renal failure, acute stroke, and transplant rejection. In this review, the authors discuss the state of the art regarding the status of drug discovery efforts targeting S1P receptors and place emphasis on potential clinical applications. Key Words: Sphingosine 1-phosphate, G protein-coupled receptor, Fingolimod, FTY720, Drug discovery, S1P agonist GPCR IN DRUG DISCOVERY include Claritin a H1 histamine receptor antagonist, Cozaar an AT1 angiotensin receptor antagonist, Neurontin a GABAB G protein-coupled receptors (GPCRs) constitute the larg- γ-aminobutyric acid receptor agonist, Plavix a P2Y12 ADP re- est superfamily of receptors for signaling molecules, ligands, ceptor antagonist, Singulair a CysLT1 leukotriene D4 receptor and currently comprise some 865 receptors (Im, 2002, 2013; antagonist, Zantac a H2 histamine receptor antagonist, and Fredriksson et al., 2003; Kihara et al., 2015). GPCRs are also Zyprexa a mixed D2/D1/5-HT2 dopamine/serotonin receptors known as 7TM receptors, because they have seven trans- antagonist. About 55 GPCRs have been cloned and identified membrane domains. These receptors may signal though G as receptors for intercellular lipid mediators, and recent studies proteins but they also initiate signals via other entities (Dav- have unearthed their functional roles under both physiological enport et al., 2013). Many ligands, including hormones, au- and pathological conditions (Im, 2004, 2009, 2013). Further- tacoids, neurotransmitters, and very small molecules to large more, the number of drug discovery studies being conducted proteins can bind and activate GPCRs, and their activations on GPCRs have increased in many fields including cancer, lead to a multitude of physiological processes (Howard et al., cardiac dysfunction, central nervous system disorders, inflam- 2001; Overington et al., 2006; Im, 2013). matory diseases, metabolic disorders, and obesity, (Im, 2004, GPCRs represent a major drug target in all clinical areas. 2009; Mutoh et al., 2012; Pyne et al., 2012; Choi and Chun, Currently, about 40% of drugs on the market target GPCRs 2013; Makide et al., 2014; Proia and Hla, 2015). Here, we and regulate their activities positively or negatively, because summarize current knowledge on one specific aspect of drug a variety of GPCRs offer selectivity and specificity for many discovery involving interactions between GPCRs and the in- human diseases (Im, 2013). Examples of their applications tercellular lipid mediator, sphingosine 1 phosphate (S1P). Open Access https://doi.org/10.4062/biomolther.2016.160 Received Jul 22, 2016 Revised Oct 6, 2016 Accepted Oct 27, 2016 Published Online Jan 1, 2017 This is an Open Access article distributed under the terms of the Creative Com- mons Attribution Non-Commercial License (http://creativecommons.org/licens- *Corresponding Author es/by-nc/4.0/) which permits unrestricted non-commercial use, distribution, E-mail: [email protected] and reproduction in any medium, provided the original work is properly cited. Tel: +82-51-510-2817, Fax: +82-51-513-6754 Copyright © 2017 The Korean Society of Applied Pharmacology www.biomolther.org 80 Park and Im. S1P Receptor Modulators and Drug Discovery SPHINGOSINE 1-PHOSPHATE AND ITS GPCRs disease-modifying drug to treat relapses of multiple sclerosis (Kihara et al., 2015). Multiple sclerosis is a demyelinating dis- S1P is a bioactive lysophospholipid metabolite that can act ease that damages axonal myelin sheaths in the brain and as an intercellular lipid mediator (Moolenaar and Hla, 2012). spinal cord. The primary action mechanism of fingolimod is to Initially, S1P was reported to be a second messenger that reduce lymphocyte egress from secondary lymphoid organ, mediates increases in intracellular calcium levels as a result thymus, and bone marrow, resulting in lymphopenia (Adachi of PDGF and IgE signaling (Olivera and Spiegel, 1993; Choi and Chiba, 2008). Thereby, lymphopenia contributes to inhibit et al., 1996). However, S1P was also unexpectedly found to axon myelin sheath damage. function as an intercellular first messenger like autacoids. The Fingolimod is a unique drug in two respects. First, fingolimod presence of S1P GPCRs in the plasma membrane was sug- is a prodrug (Fig. 1). In vivo fingolimod is phosphorylated by gested by the pertussis toxin sensitivity in S1P-induced actions sphingosine kinases (SK1 and SK2) and then phosphorylated (Bunemann et al., 1995; Goodemote et al., 1995; Im et al., fingolimod acts as an agonist on four S1P receptor subtypes 1997; Okajima et al., 1997; van Koppen et al., 1996). The dis- (S1P1, S1P3, S1P4 and S1P5) (Billich et al., 2003). Second, covery of S1P1 (formerly known as Edg-1) in 1998 along with effects of fingolimod on lymphocyte egress involve not ago- four other S1P2-5 receptors represents a milestone in sphingo- nistic but rather functional antagonistic activity against S1P1 lipid biology (An et al., 1997; Lee et al., 1998; Lynch and Im, (Fig. 1) (Matloubian et al., 2004). Both S1P and fingolimod- 1999; Okamoto et al., 1999; Im et al., 2000; Van Brocklyn et phosphate have been reported to induce lymphopenia via the al., 2000; Yamazaki et al., 2000), and leads to the identifica- agonistic activation of S1P1 and subsequent internalization of tion of a variety of biological functions mediated by interactions S1P1 in the lymphocytes (Brinkmann et al., 2004; Thangada between S1P and S1P receptors (Sanchez and Hla, 2004). In et al., 2010). In fact, the absence of S1P1 on the cell surface particular, S1P regulates the response and function of various blocks re-circulation of lymphocytes from secondary lymphoid cellular and organ systems, such as, cell differentiation, cell organs to blood, because lymphocytes egress by chemotactic migration, cell proliferation, immune response, trafficking of T response to S1P concentration gradient (high in blood and low and B cells, and vascular stability (Gardell et al., 2006; Huwiler in lymph node) through S1P1 (Brinkmann et al., 2004; Chiba, and Pfeilschifter, 2008). S1P receptors exhibit overlapping or 2009). In the case of S1P1 agonist like S1P, internalized S1P1 distinct expression patterns in various cells and tissues, and recycled back to the cell surface within several hours. Howev- as a result, the various cellular functions of S1P have been er, in the case of S1P1 modulators like fingolimod-phosphate, assigned to S1P receptor subtypes. Furthermore, because internalized S1P1 undergoes proteosomal degradation, result- S1P plays critical roles in autoimmune diseases, cancer, and ing in long-time absence of S1P1 until de novo synthesized diseases related to the cardiovascular, immune, nervous, and (Oo et al., 2007). This kind of functional antagonism of fingo- reproductive systems, S1P receptors become important treat- limod means it has a long action time, which can sometimes ment targets (Kihara et al., 2015). In addition, the discovery of be disadvantageous (Subei and Cohen, 2015). The immune S1P receptors made the screening and development of S1P modulatory action of fingolimod has also been reported in agonists and antagonists accessible. Furthermore, discover- autoimmune diseases other than multiple sclerosis, such as ies of S1P receptor subtype-selective agonists or antagonists spontaneous autoimmnune polyneuropathy and experimental could provide novel therapeutic candidates (Im, 2010). autoimmune neuritis (Kim et al., 2009; Zhang et al., 2009a). The discovery that fingolimod (also known as FTY720) is Other applications of fingolimod have also been suggested, an agonist of four S1P receptor subtypes by researchers in im- such as, for the treatment of ischemia/reperfusion injury, which mune therapeutics field accelerated the drug discovery in this is the cellular damage that results from ischemia and re-sup- area. Fingolimod has been approved as a first-in-class drug ply of blood to infarcted tissues. In fact, it has been estimated targeting S1P1 and several selective S1P receptor modulators that systemic inflammatory response after ischemia/reperfu-
Recommended publications
  • Retinamide Increases Dihydroceramide and Synergizes with Dimethylsphingosine to Enhance Cancer Cell Killing
    2967 N-(4-Hydroxyphenyl)retinamide increases dihydroceramide and synergizes with dimethylsphingosine to enhance cancer cell killing Hongtao Wang,1 Barry J. Maurer,1 Yong-Yu Liu,2 elevations in dihydroceramides (N-acylsphinganines), Elaine Wang,3 Jeremy C. Allegood,3 Samuel Kelly,3 but not desaturated ceramides, and large increases in Holly Symolon,3 Ying Liu,3 Alfred H. Merrill, Jr.,3 complex dihydrosphingolipids (dihydrosphingomyelins, Vale´rie Gouaze´-Andersson,4 Jing Yuan Yu,4 monohexosyldihydroceramides), sphinganine, and sphin- Armando E. Giuliano,4 and Myles C. Cabot4 ganine 1-phosphate. To test the hypothesis that elevation of sphinganine participates in the cytotoxicity of 4-HPR, 1Childrens Hospital Los Angeles, Keck School of Medicine, cells were treated with the sphingosine kinase inhibitor University of Southern California, Los Angeles, California; D-erythro-N,N-dimethylsphingosine (DMS), with and 2 College of Pharmacy, University of Louisiana at Monroe, without 4-HPR. After 24 h, the 4-HPR/DMS combination Monroe, Louisiana; 3School of Biology and Petit Institute of Bioengineering and Bioscience, Georgia Institute of Technology, caused a 9-fold increase in sphinganine that was sustained Atlanta, Georgia; and 4Gonda (Goldschmied) Research through +48 hours, decreased sphinganine 1-phosphate, Laboratories at the John Wayne Cancer Institute, and increased cytotoxicity. Increased dihydrosphingolipids Saint John’s Health Center, Santa Monica, California and sphinganine were also found in HL-60 leukemia cells and HT-29 colon cancer cells treated with 4-HPR. The Abstract 4-HPR/DMS combination elicited increased apoptosis in all three cell lines. We propose that a mechanism of N Fenretinide [ -(4-hydroxyphenyl)retinamide (4-HPR)] is 4-HPR–induced cytotoxicity involves increases in dihy- cytotoxic in many cancer cell types.
    [Show full text]
  • Neurological S1P Signaling As an Emerging Mechanism of Action of Oral FTY720 (Fingolimod) in Multiple Sclerosis
    Arch Pharm Res Vol 33, No 10, 1567-1574, 2010 DOI 10.1007/s12272-010-1008-5 REVIEW Neurological S1P Signaling as an Emerging Mechanism of Action of Oral FTY720 (Fingolimod) in Multiple Sclerosis Chang Wook Lee1, Ji Woong Choi2, and Jerold Chun1 1Department of Molecular Biology, Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, California 92037, USA and 2Department of Pharmacology, Division of Basic Medicine and Science, Gachon University of Medi- cine and Science, Incheon 406-799, Korea (Received August 15, 2010/Revised August 23, 2010/Accepted August 24, 2010) FTY720 (fingolimod, Novartis) is a promising investigational drug for relapsing forms of multi- ple sclerosis (MS), an autoimmune and neurodegenerative disorder of the central nervous sys- tem. It is currently under FDA review in the United States, and could represent the first approved oral treatment for MS. Extensive, ongoing clinical trials in Phase II/III have sup- ported both the efficacy and safety of FTY720. FTY720 itself is not bioactive, but when phospho- rylated (FTY720-P) by sphingosine kinase 2, it becomes active through modulation of 4 of the 5 known G protein-coupled sphingosine 1-phosphate (S1P) receptors. The mechanism of action (MOA) is thought to be immunological, where FTY720 alters lymphocyte trafficking via S1P1. However, MOA for FTY720 in MS may also involve a direct, neurological action within the cen- tral nervous system in view of documented S1P receptor-mediated signaling influences in the brain, and this review considers observations that support an emerging neurological MOA. Key words: FTY720, Fingolimod, Sphingosine 1-phosphate receptors, Multiple sclerosis, Cen- tral nervous system INTRODUCTION factors that initiate MS are still unknown.
    [Show full text]
  • Fingolimod (FTY720): Discovery and Development of an Oral Drug to Treat Multiple Sclerosis
    REVIEWS CASE HISTORY Fingolimod (FTY720): discovery and development of an oral drug to treat multiple sclerosis Volker Brinkmann*, Andreas Billich*, Thomas Baumruker*, Peter Heining‡, Robert Schmouder§, Gordon Francis||, Shreeram Aradhye¶ and Pascale Burtin# Abstract | The discovery of fingolimod (FTY720/Gilenya; Novartis), an orally active immunomodulatory drug, has opened up new approaches to the treatment of multiple sclerosis, the most common inflammatory disorder of the central nervous system. Elucidation of the effects of fingolimod — mediated by the modulation of sphingosine 1‑phosphate (S1P) receptors — has indicated that its therapeutic activity could be due to regulation of the migration of selected lymphocyte subsets into the central nervous system and direct effects on neural cells, particularly astrocytes. An improved understanding of the biology of S1P receptors has also been gained. This article describes the discovery and development of fingolimod, which was approved by the US Food and Drug Administration in September 2010 as a first‑line treatment for relapsing forms of multiple sclerosis, thereby becoming the first oral disease‑modifying therapy to be approved for multiple sclerosis in the United States. Demyelination Multiple sclerosis (MS) is a chronic autoimmune disorder Treatment strategies for MS usually involve the man- Damage of the myelin sheath of the central nervous system (CNS) that is characterized agement of symptoms and the use of disease-modifying of axons. A demyelinating by inflammation leading to astrogliosis, demyelination, drugs to reduce the frequency of relapses and to slow disease is any disease of and loss of oligodendrocytes and neurons1. MS is the the progression of disability. Established first-line the nervous system in which the myelin sheath is damaged.
    [Show full text]
  • Targeting the Sphingosine Kinase/Sphingosine-1-Phosphate Signaling Axis in Drug Discovery for Cancer Therapy
    cancers Review Targeting the Sphingosine Kinase/Sphingosine-1-Phosphate Signaling Axis in Drug Discovery for Cancer Therapy Preeti Gupta 1, Aaliya Taiyab 1 , Afzal Hussain 2, Mohamed F. Alajmi 2, Asimul Islam 1 and Md. Imtaiyaz Hassan 1,* 1 Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, Jamia Nagar, New Delhi 110025, India; [email protected] (P.G.); [email protected] (A.T.); [email protected] (A.I.) 2 Department of Pharmacognosy, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia; afi[email protected] (A.H.); [email protected] (M.F.A.) * Correspondence: [email protected] Simple Summary: Cancer is the prime cause of death globally. The altered stimulation of signaling pathways controlled by human kinases has often been observed in various human malignancies. The over-expression of SphK1 (a lipid kinase) and its metabolite S1P have been observed in various types of cancer and metabolic disorders, making it a potential therapeutic target. Here, we discuss the sphingolipid metabolism along with the critical enzymes involved in the pathway. The review provides comprehensive details of SphK isoforms, including their functional role, activation, and involvement in various human malignancies. An overview of different SphK inhibitors at different phases of clinical trials and can potentially be utilized as cancer therapeutics has also been reviewed. Citation: Gupta, P.; Taiyab, A.; Hussain, A.; Alajmi, M.F.; Islam, A.; Abstract: Sphingolipid metabolites have emerged as critical players in the regulation of various Hassan, M..I. Targeting the Sphingosine Kinase/Sphingosine- physiological processes. Ceramide and sphingosine induce cell growth arrest and apoptosis, whereas 1-Phosphate Signaling Axis in Drug sphingosine-1-phosphate (S1P) promotes cell proliferation and survival.
    [Show full text]
  • Gilenya; FTY720
    STRADER, CHERILYN R., M.S. Fingolimod (Gilenya; FTY720): A Recently Approved Multiple Sclerosis Drug Based on a Fungal Secondary Metabolite and the Creation of a Natural Products Pure Compound Database and Organized Storage System. (2012) Directed by Dr. Nicholas H. Oberlies, 78 pp. Fingolimod (Gilenya; FTY720), a synthetic compound based on the fungal secondary metabolite, myriocin (ISP-I), is a potent immunosuppressant that was approved in September 2010 by the U.S. FDA as a new treatment for multiple sclerosis (MS). Fingolimod was synthesized by the research group of Tetsuro Fujita at Kyoto University in 1992 while investigating structure-activity relationships of derivatives of the fungal metabolite, ISP-I, isolated from Isaria sinclairii. Fingolimod becomes active in vivo following phosphorylation by sphingosine kinase 2 to form fingolimod-phosphate, which binds to specific G protein-coupled receptors (GPCRs) and prevents the release of lymphocytes from lymphoid tissue. Fingolimod is orally active, which is unique among current first-line MS therapies, and it has the potential to be used in the treatment of organ transplants and cancer. The first chapter reviews the discovery and development of fingolimod, from an isolated lead natural product, through synthetic analogues, to an approved drug. Natural products play an important role in the pharmaceutical industry, accounting for approximately half of all drugs approved in the U.S. between 1981 and 2006. Given the importance of natural product research and the vast amount of data that is generated in the process, it is imperative that the pure compounds and data are stored in a manner that will allow researchers to derive as much value as possible.
    [Show full text]
  • FTY720-P) Nor SET Dimerization Or Ceramide Induction but Depends on Interaction with SET K209
    Antagonistic activities of the immunomodulator and protein phosphatase 2A (PP2A)-activating drug FTY720 (Fingolimod, Gilenya) in Jak2-driven myeloproliferative neoplasms. DISSERTATION Presented in Partial Fulfillment of the Requirements for the degree Doctor of Philosophy in the Graduate School of The Ohio State University By Joshua John Oaks, B.S. Integrated Biomedical Sciences Graduate Program The Ohio State University 2013 Dissertation Committee Danilo Perrotti MD, PhD Roger Briesewitz, PhD Denis Guttridge, PhD Harold Fisk, PhD Copyright 2013 Joshua John Oaks ABSTRACT FTY720 (Fingolimod, Gilenya) is a sphingosine analog used as an immunosuppressant in multiple sclerosis patients. FTY720 is also a potent protein phosphatase 2A (PP2A)- activating drug (PAD). PP2A is a tumor suppressor found inactivated in different types of cancer. We show here that PP2A is inactive in Polycythemia Vera (PV) and other myeloproliferative neoplasms (MPNs) characterized by the expression of the transforming Jak2V617F oncogene. PP2A inactivation occurs in a Jak2V617F dose/kinase- dependent manner through the PI-3Kγ-PKC-induced phosphorylation of the PP2A inhibitor SET and by modulation of nitric oxide synthase (NOS2)-induced nitrosylation of PP2A. Genetic and/or PAD-mediated PP2A reactivation induces Jak2V617F inactivation/downregulation and impairs clonogenic potential of Jak2V617F cell lines and PV but not normal CD34+ progenitors. Likewise, FTY720 decreases leukemic allelic burden, reduces splenomegaly and significantly increases survival of Jak2V617F
    [Show full text]
  • The Development of Fingolimod (FTY720, Gilenya)
    Discovery Medicine ® www.discoverymedicine.com n ISSN: 1539-6509; eISSN: 1944-7930 A Mechanistically Novel, First Oral Therapy for Multiple Sclerosis: The Development of Fingolimod (FTY720, Gilenya) Jerold ChuN ANd Volker BrINkmANN Abstract: Multiple sclerosis (MS) is a chronic Introduction autoimmune disorder affecting the central nervous system (CNS) through demyelination and neurode - Multiple sclerosis (MS) is a neurological disorder char - generation. Until recently, major therapeutic treat - acterized by demyelination and neurodegeneration ments have relied on agents requiring injection within the central nervous system (CNS) with common delivery. In September 2010, fingolimod/FTY720 relapsing forms produced through autoimmune inflam - (Gilenya, Novartis) was approved by the FDA as the matory mechanisms (Compston and Coles, 2002) first oral treatment for relapsing forms of MS. involving autoreactive lymphocytes that penetrate the Fingolimod is a novel compound produced by chem - blood-brain barrier to attack the nervous system (Figure ical modification of a fungal precursor. Its active 1). MS is a major cause of nervous system disability metabolite, formed by in vivo phosphorylation, mod - affecting individuals in the prime of life with a 2-3:1 ulates sphingosine 1-phosphate (S1P) receptors that female to male ratio (Compston and Coles, 2002) and are a subset of a larger family of cell-surface, G pro - an estimated global prevalence of 2.5 million that is tein-coupled receptors (GPCRs) mediating the highest in Northern latitudes particularly amongst effects of bioactive lipids known as lysophospho - Caucasians (Noseworthy et al. , 2000; Rosati, 2001). lipids. Fingolimod’s mechanism of action in MS is The initiating causes of MS remain unknown not completely understood; however, its relevant (Baranzini et al.
    [Show full text]
  • Sphingolipids in Inflammation: from Bench to Bedside
    Mediators of Inflammation Sphingolipids in Inflammation: From Bench to Bedside Guest Editors: Kazuyuki Kitatani, Kazuhisa Iwabuchi, Ashley Snider, and Laura Riboni Sphingolipids in Inflammation: From Bench to Bedside Mediators of Inflammation Sphingolipids in Inflammation: From Bench to Bedside Guest Editors: Kazuyuki Kitatani, Kazuhisa Iwabuchi, Ashley Snider, and Laura Riboni Copyright © 2016 Hindawi Publishing Corporation. All rights reserved. This is a special issue published in “Mediators of Inflammation.” All articles are open access articles distributed under the Creative Com- mons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited. Editorial Board Anshu Agrawal, USA Christoph Garlichs, Germany Eeva Moilanen, Finland Muzamil Ahmad, India Mirella Giovarelli, Italy Jonas Mudter, Germany Simi Ali, UK Denis Girard, Canada Hannes Neuwirt, Austria Amedeo Amedei, Italy Ronald Gladue, USA Marja Ojaniemi, Finland Jagadeesh Bayry, France Hermann Gram, Switzerland Sandra Helena Penha Oliveira, Brazil Philip Bufler, Germany Oreste Gualillo, Spain Vera L. Petricevich, Mexico Elisabetta Buommino, Italy Elaine Hatanaka, Brazil Carolina T. Piñeiro, Spain Luca Cantarini, Italy Nina Ivanovska, Bulgaria Marc Pouliot, Canada Claudia Cocco, Italy Yona Keisari, Israel Michal A. Rahat, Israel Dianne Cooper, UK Alex Kleinjan, Netherlands Alexander Riad, Germany Jose Crispin, Mexico Magdalena Klink, Poland Sunit K. Singh, India Fulvio D’Acquisto, UK Marije I. Koenders, Netherlands HelenC.Steel,SouthAfrica Pham My-Chan Dang, France Elzbieta Kolaczkowska, Poland Dennis D. Taub, USA Wilco de Jager, Netherlands Dmitri V. Krysko, Belgium Kathy Triantafilou, UK Beatriz De las Heras, Spain Philipp M. Lepper, Germany Fumio Tsuji, Japan Chiara De Luca, Germany Changlin Li, USA Giuseppe Valacchi, Italy Clara Di Filippo, Italy Eduardo López-Collazo, Spain Luc Vallières, Canada Maziar Divangahi, Canada Antonio Macciò, Italy Elena Voronov, Israel Amos Douvdevani, Israel A.
    [Show full text]
  • The Sphingosine 1-Phosphate Signaling Pathway in Epilepsy: a Possible Role for The
    The sphingosine 1-phosphate signaling pathway in epilepsy: a possible role for the immunomodulator drug fingolimod in epilepsy treatment 1Antonio Leo, 1Rita Citraro, 2Rosario Marra, 1Ernesto Palma, 1Eugenio Donato Di Paola, 3Andrew Constanti, 1Giovambattista De Sarro, 1, *Emilio Russo. 1Science of Health Department, School of Medicine, University “Magna Graecia” of Catanzaro, Italy; 2National Council of Research (CNR), Institute of Neurological Science, Catanzaro, Italy; 3Department of Pharmacology, UCL School of Pharmacy, 29/39 Brunswick Square, London, United Kingdom. *Author for correspondence: Prof. Emilio Russo, Ph.D., Chair of Pharmacology, Department of Science of Health, School of Medicine, University of Catanzaro, Italy Via T. Campanella, 115 88100 Catanzaro, ITALY. Phone +39 0961 3694191; Fax +39 0961 3694192 e-mail: [email protected] 1 Abstract It is currently known that erythrocytes are the major source of sphingosine 1-phosphate (S1P) in the body. S1P acts both extracellularly as a cellular mediator and intracellularly as an important second messenger molecule. Its effects are mediated by interaction with five specific types of G protein-coupled S1P receptor. Fingolimod, is a recognized modulator of S1P receptors, and is the first orally active disease-modifying therapy that has been approved for the treatment of multiple sclerosis. Magnetic resonance imaging data suggest that fingolimod may be effective in multiple sclerosis by preventing blood-brain barrier disruption and brain atrophy. Fingolimod might also possess S1P receptor-independent effects and exerts both anti- inflammatory and neuroprotective effects. In the therapeutic management of epilepsy, there are a great number of antiepileptic drugs, but there is still a need for others that are more effective and safer.
    [Show full text]
  • Uptake of Exogenous Serine Is Important to Maintain Sphingolipid
    bioRxiv preprint doi: https://doi.org/10.1101/2020.03.30.016220; this version posted March 31, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license. 1 Uptake of exogenous serine is important to maintain sphingolipid 2 homeostasis in Saccharomyces cerevisiae 3 4 Bianca M. Esch1, Sergej Limar1, André Bogdanowski2,3, Christos Gournas4,#a, 5 Tushar More5, Celine Sundag1, Stefan Walter6, Jürgen J. Heinisch7, Christer S. 6 Ejsing5,8, Bruno André4 and Florian Fröhlich1,6* 7 8 1Department of Biology/Chemistry, Molecular Membrane Biology Group, University of 9 Osnabrück, Barbarastraße 13, 49076 Osnabrück, Germany 10 11 2Department of Biology/Chemistry, Ecology Group, University of Osnabrück, Barbarastraße 12 13, 49076 Osnabrück, Germany 13 14 3UFZ–Helmholtz Centre for Environmental Research Ltd, Department for Ecological 15 Modelling, Permoserstr. 15, 04318, Leipzig, Germany 16 17 4Molecular Physiology of the Cell, Université Libre de Bruxelles (ULB), Institut de Biologie et 18 de Médecine Moléculaires, 6041 Gosselies, Belgium 19 20 5Department of Biochemistry and Molecular Biology, Villum Center for Bioanalytical 21 Sciences, University of Southern Denmark, 5000 Odense, Denmark 22 23 6Center of Cellular Nanoanalytics Osnabrück, Barbarastraße 11, 49076 Osnabrück, 24 Germany 25 26 7Department of Biology/Chemistry, Division of Genetics, University of Osnabrück, 27 Barbarastraße 11, 49076 Osnabrück, Germany 28 29 8Cell Biology and Biophysics Unit, European Molecular Biology Laboratory, 69117 30 Heidelberg, Germany 31 32 33 *Corresponding author: 34 Email: [email protected] 35 Phone: +49-541-969-2961 36 37 38 #aCurrent address: 39 Microbial Molecular Genetics Laboratory 40 Institute of Biosciences and Applications (IBA) 41 National Centre for Scientific Research “Demokritos” (NCSRD) 42 Patr.
    [Show full text]
  • Regulation of the Activity of Mammalian Serine Palmitoyltransferase
    Zurich Open Repository and Archive University of Zurich Main Library Strickhofstrasse 39 CH-8057 Zurich www.zora.uzh.ch Year: 2016 Regulation of the activity of mammalian serine palmitoyltransferase Zhakupova, Assem Posted at the Zurich Open Repository and Archive, University of Zurich ZORA URL: https://doi.org/10.5167/uzh-129376 Dissertation Published Version Originally published at: Zhakupova, Assem. Regulation of the activity of mammalian serine palmitoyltransferase. 2016, Univer- sity of Zurich, Faculty of Science. Regulation of the Activity of Mammalian Serine Palmitoyltransferase ___________________________________________________________________________ Dissertation zur Erlangung der naturwissenschaftlichen Doktorwürde (Dr. sc. nat.) vorgelegt der Mathematisch-naturwissenschaftlichen Fakultät der Universität Zürich von Assem Zhakupova aus Kasachstan Promotionskomitee Prof. Dr. Arnold von Eckardstein (Vorsitz) Prof. Dr. Thorsten Hornemann Prof. Dr. Stephan Neuhauss PD Dr. Sabrina Sonda Dr. Ludovic Gillet Zürich, 2016 ii Table of contents Abstract............................................................................................................................................iv Zusammenfassung……………………………………………………………………………………………….vi General Introduction……………………………………………………………………………………………1 I. The sphingolipid metabolic pathway…………………………………………………………….1 II. The sphingolipid salvage pathway………………………………………………………………...5 III. Intracellular sphingolipid trafficking and transport……………………………………….7 IV. The mammalian SPT complex……………………………………………………………………….8
    [Show full text]
  • Effects of Inhibition of Serine Palmitoyltransferase (SPT) and Sphingosine Kinase 1 (Sphk1) on Palmitate Induced Insulin Resistance in L6 Myotubes
    Effects of Inhibition of Serine Palmitoyltransferase (SPT) and Sphingosine Kinase 1 (SphK1) on Palmitate Induced Insulin Resistance in L6 Myotubes Agnieszka Mikłosz*, Bartłomiej Łukaszuk, Marcin Baranowski, Jan Górski, Adrian Chabowski Department of Physiology, Medical University of Bialystok, Bialystok, Poland Abstract Background: The objective of this study was to examine the effects of short (2 h) and prolonged (18 h) inhibition of serine palmitoyltransferase (SPT) and sphingosine kinase 1 (SphK1) on palmitate (PA) induced insulin resistance in L6 myotubes. Methods: L6 myotubes were treated simultaneously with either PA and myriocin (SPT inhibitor) or PA and Ski II (SphK1inhibitor) for different time periods (2 h and 18 h). Insulin stimulated glucose uptake was measured using radioactive isotope. Expression of insulin signaling proteins was determined using Western blot analyses. Intracellular sphingolipids content [sphinganine (SFA), ceramide (CER), sphingosine (SFO), sphingosine-1- phosphate (S1P)] were estimated by HPLC. Results: Our results revealed that both short and prolonged time of inhibition of SPT by myriocin was sufficient to prevent ceramide accumulation and simultaneously reverse palmitate induced inhibition of insulin-stimulated glucose transport. In contrast, prolonged inhibition of SphK1 intensified the effect of PA on insulin-stimulated glucose uptake and attenuated further the activity of insulin signaling proteins (pGSK3β/GSK3β ratio) in L6 myotubes. These effects were related to the accumulation of sphingosine in palmitate treated myotubes. Conclusion: Myriocin is more effective in restoration of palmitate induced insulin resistance in L6 myocytes, despite of the time of SPT inhibition, comparing to SKII (a specific SphK1 inhibitor). Observed changes in insulin signaling proteins were related to the content of specific sphingolipids, namely to the reduction of ceramide.
    [Show full text]