1
|
Miyamoto K, Sujino T, Harada Y, Ashida H, Yoshimatsu Y, Yonemoto Y, Nemoto Y, Tomura M, Melhem H, Niess JH, Suzuki T, Suzuki T, Suzuki S, Koda Y, Okamoto R, Mikami Y, Teratani T, Tanaka KF, Yoshimura A, Sato T, Kanai T. The gut microbiota-induced kynurenic acid recruits GPR35-positive macrophages to promote experimental encephalitis. Cell Rep 2023; 42:113005. [PMID: 37590143 DOI: 10.1016/j.celrep.2023.113005] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2023] [Revised: 07/25/2023] [Accepted: 08/02/2023] [Indexed: 08/19/2023] Open
Abstract
The intricate interplay between gut microbes and the onset of experimental autoimmune encephalomyelitis (EAE) remains poorly understood. Here, we uncover remarkable similarities between CD4+ T cells in the spinal cord and their counterparts in the small intestine. Furthermore, we unveil a synergistic relationship between the microbiota, particularly enriched with the tryptophan metabolism gene EC:1.13.11.11, and intestinal cells. This symbiotic collaboration results in the biosynthesis of kynurenic acid (KYNA), which modulates the recruitment and aggregation of GPR35-positive macrophages. Subsequently, a robust T helper 17 (Th17) immune response is activated, ultimately triggering the onset of EAE. Conversely, modulating the KYNA-mediated GPR35 signaling in Cx3cr1+ macrophages leads to a remarkable amelioration of EAE. These findings shed light on the crucial role of microbial-derived tryptophan metabolites in regulating immune responses within extraintestinal tissues.
Collapse
Affiliation(s)
- Kentaro Miyamoto
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan; Miyarisan Pharmaceutical Co., Ltd., Research Laboratory, 1-10-3, Kaminagazato, Kita-ku, Tokyo 114-0016, Japan
| | - Tomohisa Sujino
- Center for Diagnostic and Therapeutic Endoscopy, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan.
| | - Yosuke Harada
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Hiroshi Ashida
- Department of Bacterial Infection and Host Response, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), 1-5-45, Yushima, Bunkyo-ku, Tokyo 113-8510, Japan; Medical Mycology Research Center, Chiba University, 1-8-1, Inohana, Cyuo-ku, Chiba city, Chiba 260-8673, Japan
| | - Yusuke Yoshimatsu
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Yuki Yonemoto
- Department of Gastroenterology and Hepatology, Tokyo Medical Dental University (TMDU), 1-5-45, Yushima, Bunkyo-ku, Tokyo 113-8510, Japan
| | - Yasuhiro Nemoto
- Department of Gastroenterology and Hepatology, Tokyo Medical Dental University (TMDU), 1-5-45, Yushima, Bunkyo-ku, Tokyo 113-8510, Japan
| | - Michio Tomura
- Laboratory of Immunology, Faculty of Pharmacy, Osaka Otani University, 3-11-1 Nshikiorikita, Tondabayshi, Osaka, 584-8584, Japan
| | - Hassan Melhem
- Department of Biomedicine, University of Basel, 4031 Basel, Switzerland
| | - Jan Hendrik Niess
- Department of Biomedicine, University of Basel, 4031 Basel, Switzerland; Clarunis-University Center for Gastrointestinal and Liver Diseases, University Hospital Basel, 4002 Basel, Switzerland
| | - Toshihiko Suzuki
- Department of Bacterial Infection and Host Response, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), 1-5-45, Yushima, Bunkyo-ku, Tokyo 113-8510, Japan
| | - Toru Suzuki
- Division of Brain Sciences Institute for Advanced Medical Research, Keio University School of Medicne, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Shohei Suzuki
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Yuzo Koda
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Ryuichi Okamoto
- Department of Gastroenterology and Hepatology, Tokyo Medical Dental University (TMDU), 1-5-45, Yushima, Bunkyo-ku, Tokyo 113-8510, Japan
| | - Yohei Mikami
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Toshiaki Teratani
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Kenji F. Tanaka
- Division of Brain Sciences Institute for Advanced Medical Research, Keio University School of Medicne, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Akihiko Yoshimura
- Department of Microbiology and Immunology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Toshiro Sato
- Department of Organoid Medicine, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Takanori Kanai
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan; AMED-CREST, Japan Agency for Medical Research and Development, 1-7-1, Otemachi, Chiyoda-ku, Tokyo 100-0004, Japan.
| |
Collapse
|
2
|
Cha E, Kim J, Gotina L, Kim J, Kim HJ, Seo SH, Park JE, Joo J, Kang M, Lee J, Hwang H, Kim HJ, Pae AN, Park KD, Park JH, Lim SM. Exploration of Tetrahydroisoquinoline- and Benzo[ c]azepine-Based Sphingosine 1-Phosphate Receptor 1 Agonists for the Treatment of Multiple Sclerosis. J Med Chem 2023; 66:10381-10412. [PMID: 37489798 DOI: 10.1021/acs.jmedchem.3c00498] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/26/2023]
Abstract
Because of the wide use of Fingolimod for the treatment of multiple sclerosis (MS) and its cardiovascular side effects such as bradycardia, second-generation sphingosine 1-phosphate receptor 1 (S1P1) agonist drugs for MS have been developed and approved by FDA. The issue of bradycardia is still present with the new drugs, however, which necessitates further exploration of S1P1 agonists with improved safety profiles for next-generation MS drugs. Herein, we report a tetrahydroisoquinoline or a benzo[c]azepine core-based S1P1 agonists such as 32 and 60 after systematic examination of hydrophilic groups and cores. We investigated the binding modes of our representative compounds and their molecular interactions with S1P1 employing recent S1P1 cryo-EM structures. Also, favorable ADME properties of our compounds were shown. Furthermore, in vivo efficacy of our compounds was clearly demonstrated with PLC and EAE studies. Also, the preliminary in vitro cardiovascular safety of our compound was verified with human iPSC-derived cardiomyocytes.
Collapse
Affiliation(s)
- Eunji Cha
- Center for Brain Disorders, Brain Science Institute, Korea Institute of Science and Technology, 5, Hwarang-ro 14-gil, Seongbuk-gu, Seoul 02792, Republic of Korea
- Department of Chemistry, Korea University, 145 Anam-ro, Seongbuk-gu, Seoul 02841, Republic of Korea
| | - Jushin Kim
- Center for Brain Disorders, Brain Science Institute, Korea Institute of Science and Technology, 5, Hwarang-ro 14-gil, Seongbuk-gu, Seoul 02792, Republic of Korea
- Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, 50, Yonsei-ro, Seodaemun-gu, Seoul 03722, Republic of Korea
| | - Lizaveta Gotina
- Center for Brain Disorders, Brain Science Institute, Korea Institute of Science and Technology, 5, Hwarang-ro 14-gil, Seongbuk-gu, Seoul 02792, Republic of Korea
- Division of Bio-Medical Science & Technology, KIST School, University of Science and Technology, 5, Hwarang-ro 14-gil, Seongbuk-gu, Seoul 02792, Republic of Korea
| | - Jaehwan Kim
- Center for Brain Disorders, Brain Science Institute, Korea Institute of Science and Technology, 5, Hwarang-ro 14-gil, Seongbuk-gu, Seoul 02792, Republic of Korea
- Division of Bio-Medical Science & Technology, KIST School, University of Science and Technology, 5, Hwarang-ro 14-gil, Seongbuk-gu, Seoul 02792, Republic of Korea
| | - Hyeon Jeong Kim
- Center for Brain Disorders, Brain Science Institute, Korea Institute of Science and Technology, 5, Hwarang-ro 14-gil, Seongbuk-gu, Seoul 02792, Republic of Korea
- Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, 50, Yonsei-ro, Seodaemun-gu, Seoul 03722, Republic of Korea
| | - Seon Hee Seo
- Center for Brain Disorders, Brain Science Institute, Korea Institute of Science and Technology, 5, Hwarang-ro 14-gil, Seongbuk-gu, Seoul 02792, Republic of Korea
| | - Jeong-Eun Park
- New Drug Development Center, Daegu Gyeongbuk Medical Innovation Foundation, 80 Cheombok-ro, Dong-gu, Daegu 41061, Republic of Korea
| | - Jeongmin Joo
- New Drug Development Center, Daegu Gyeongbuk Medical Innovation Foundation, 80 Cheombok-ro, Dong-gu, Daegu 41061, Republic of Korea
| | - Minsik Kang
- Doping Control Center, Research Resources Division, Korea Institute of Science and Technology, 5, Hwarang-ro 14-gil, Seongbuk-gu, Seoul 02792, Republic of Korea
| | - Jaeick Lee
- Doping Control Center, Research Resources Division, Korea Institute of Science and Technology, 5, Hwarang-ro 14-gil, Seongbuk-gu, Seoul 02792, Republic of Korea
| | - Hayoung Hwang
- New Drug Development Center, Daegu Gyeongbuk Medical Innovation Foundation, 80 Cheombok-ro, Dong-gu, Daegu 41061, Republic of Korea
| | - Hak Joong Kim
- Department of Chemistry, Korea University, 145 Anam-ro, Seongbuk-gu, Seoul 02841, Republic of Korea
| | - Ae Nim Pae
- Center for Brain Disorders, Brain Science Institute, Korea Institute of Science and Technology, 5, Hwarang-ro 14-gil, Seongbuk-gu, Seoul 02792, Republic of Korea
- Division of Bio-Medical Science & Technology, KIST School, University of Science and Technology, 5, Hwarang-ro 14-gil, Seongbuk-gu, Seoul 02792, Republic of Korea
| | - Ki Duk Park
- Center for Brain Disorders, Brain Science Institute, Korea Institute of Science and Technology, 5, Hwarang-ro 14-gil, Seongbuk-gu, Seoul 02792, Republic of Korea
- Division of Bio-Medical Science & Technology, KIST School, University of Science and Technology, 5, Hwarang-ro 14-gil, Seongbuk-gu, Seoul 02792, Republic of Korea
| | - Jong-Hyun Park
- Center for Brain Disorders, Brain Science Institute, Korea Institute of Science and Technology, 5, Hwarang-ro 14-gil, Seongbuk-gu, Seoul 02792, Republic of Korea
- Division of Bio-Medical Science & Technology, KIST School, University of Science and Technology, 5, Hwarang-ro 14-gil, Seongbuk-gu, Seoul 02792, Republic of Korea
| | - Sang Min Lim
- Center for Brain Disorders, Brain Science Institute, Korea Institute of Science and Technology, 5, Hwarang-ro 14-gil, Seongbuk-gu, Seoul 02792, Republic of Korea
- Division of Bio-Medical Science & Technology, KIST School, University of Science and Technology, 5, Hwarang-ro 14-gil, Seongbuk-gu, Seoul 02792, Republic of Korea
| |
Collapse
|
3
|
Jiang ZJ, Gong LW. The SphK1/S1P Axis Regulates Synaptic Vesicle Endocytosis via TRPC5 Channels. J Neurosci 2023; 43:3807-3824. [PMID: 37185099 PMCID: PMC10217994 DOI: 10.1523/jneurosci.1494-22.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Revised: 04/13/2023] [Accepted: 04/17/2023] [Indexed: 05/17/2023] Open
Abstract
Sphingosine-1-phosphate (S1P), a bioactive sphingolipid concentrated in the brain, is essential for normal brain functions, such as learning and memory and feeding behaviors. Sphingosine kinase 1 (SphK1), the primary kinase responsible for S1P production in the brain, is abundant within presynaptic terminals, indicating a potential role of the SphK1/S1P axis in presynaptic physiology. Altered S1P levels have been highlighted in many neurologic diseases with endocytic malfunctions. However, it remains unknown whether the SphK1/S1P axis may regulate synaptic vesicle endocytosis in neurons. The present study evaluates potential functions of the SphK1/S1P axis in synaptic vesicle endocytosis by determining effects of a dominant negative catalytically inactive SphK1. Our data for the first time identify a critical role of the SphK1/S1P axis in endocytosis in both neuroendocrine chromaffin cells and neurons from mice of both sexes. Furthermore, our Ca2+ imaging data indicate that the SphK1/S1P axis may be important for presynaptic Ca2+ increases during prolonged stimulations by regulating the Ca2+ permeable TRPC5 channels, which per se regulate synaptic vesicle endocytosis. Collectively, our data point out a critical role of the regulation of TRPC5 by the SphK1/S1P axis in synaptic vesicle endocytosis.SIGNIFICANCE STATEMENT Sphingosine kinase 1 (SphK1), the primary kinase responsible for brain sphingosine-1-phosphate (S1P) production, is abundant within presynaptic terminals. Altered SphK1/S1P metabolisms has been highlighted in many neurologic disorders with defective synaptic vesicle endocytosis. However, whether the SphK1/S1P axis may regulate synaptic vesicle endocytosis is unknown. Here, we identify that the SphK1/S1P axis regulates the kinetics of synaptic vesicle endocytosis in neurons, in addition to controlling fission-pore duration during single vesicle endocytosis in neuroendocrine chromaffin cells. The regulation of the SphK1/S1P axis in synaptic vesicle endocytosis is specific since it has a distinguished signaling pathway, which involves regulation of Ca2+ influx via TRPC5 channels. This discovery may provide novel mechanistic implications for the SphK1/S1P axis in brain functions under physiological and pathologic conditions.
Collapse
Affiliation(s)
- Zhong-Jiao Jiang
- Department of Biological Sciences, University of Illinois Chicago, Chicago, Illinois 60607
| | - Liang-Wei Gong
- Department of Biological Sciences, University of Illinois Chicago, Chicago, Illinois 60607
| |
Collapse
|
4
|
Li F, Zhang Y, Lin Z, Yan L, Liu Q, Li Y, Pei X, Feng Y, Han X, Yang J, Zheng F, Li T, Zhang Y, Fu Z, Shao D, Yu J, Li C. Targeting SPHK1/S1PR3-regulated S-1-P metabolic disorder triggers autophagic cell death in pulmonary lymphangiomyomatosis (LAM). Cell Death Dis 2022; 13:1065. [PMID: 36543771 PMCID: PMC9772321 DOI: 10.1038/s41419-022-05511-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Revised: 12/07/2022] [Accepted: 12/09/2022] [Indexed: 12/24/2022]
Abstract
Lymphangioleiomyomatosis (LAM), a progressive pulmonary disease exclusively affecting females, is caused by defects or mutations in the coding gene tuberous sclerosis complex 1 (TSC1) or TSC2, causing the mammalian target of rapamycin complex 1 (mTORC1) activation and autophagy inhibition. Clinically, rapamycin shows limited cytocidal effects, and LAM recurs after drug withdrawal. In this study, we demonstrated that TSC2 negatively regulated the sphingolipid metabolism pathway and the expressions of sphingosine kinase 1 (SPHK1) and sphingosine-1-phosphate receptor 3 (S1PR3) were significantly elevated in LAM patient-derived TSC2-deficient cells compared to TSC2-addback cells, insensitive to rapamycin treatment and estrogen stimulation. Knockdown of SPHK1 showed reduced viability, migration and invasion in TSC2-deficient cells. Selective SPHK1 antagonist PF543 potently suppressed the viability of TSC2-deficient cells and induced autophagy-mediated cell death. Meanwhile, the cognate receptor S1PR3 was identified to mediating the tumorigenic effects of sphingosine-1-phosphate (S1P). Treatment with TY52156, a selective antagonist for S1PR3, or genetic silencing using S1PR3-siRNA suppressed the viability of TSC2-deficient cells. Both SPHK1 and S1PR3 inhibitors markedly exhibited antitumor effect in a xenograft model of TSC2-null cells, restored autophagy level, and triggered cell death. Together, we identified novel rapamycin-insensitive sphingosine metabolic signatures in TSC2-null LAM cells. Therapeutic targeting of aberrant SPHK1/S1P/S1PR3 signaling may have potent therapeutic benefit for patients with TSC/LAM or other hyperactive mTOR neoplasms with autophagy inhibition.
Collapse
Affiliation(s)
- Fei Li
- grid.216938.70000 0000 9878 7032State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Nankai University, 300350 Tianjin, P.R. China
| | - Yifan Zhang
- grid.216938.70000 0000 9878 7032State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Nankai University, 300350 Tianjin, P.R. China
| | - Zhoujun Lin
- grid.216938.70000 0000 9878 7032State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Nankai University, 300350 Tianjin, P.R. China
| | - Lizhong Yan
- grid.216938.70000 0000 9878 7032State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Nankai University, 300350 Tianjin, P.R. China
| | - Qiao Liu
- grid.216938.70000 0000 9878 7032State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Nankai University, 300350 Tianjin, P.R. China
| | - Yin Li
- grid.216938.70000 0000 9878 7032State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Nankai University, 300350 Tianjin, P.R. China
| | - Xiaolin Pei
- grid.216938.70000 0000 9878 7032State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Nankai University, 300350 Tianjin, P.R. China
| | - Ya Feng
- grid.216938.70000 0000 9878 7032State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Nankai University, 300350 Tianjin, P.R. China
| | - Xiao Han
- grid.216938.70000 0000 9878 7032State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Nankai University, 300350 Tianjin, P.R. China
| | - Juan Yang
- grid.216938.70000 0000 9878 7032State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Nankai University, 300350 Tianjin, P.R. China
| | - Fangxu Zheng
- grid.216938.70000 0000 9878 7032State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Nankai University, 300350 Tianjin, P.R. China
| | - Tianjiao Li
- grid.216938.70000 0000 9878 7032State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Nankai University, 300350 Tianjin, P.R. China
| | - Yupeng Zhang
- grid.216938.70000 0000 9878 7032State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Nankai University, 300350 Tianjin, P.R. China
| | - Zhenkun Fu
- grid.216938.70000 0000 9878 7032State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Nankai University, 300350 Tianjin, P.R. China ,grid.410736.70000 0001 2204 9268Department of Immunology & Wu Lien-Teh Institute & Heilongjiang Provincial Key Laboratory for Infection and Immunity, Harbin Medical University & Heilongjiang Academy of Medical Science, Harbin, China
| | - Di Shao
- grid.414287.c0000 0004 1757 967XChongqing University Central Hospital, Chongqing Emergency Medical Center, 400000 Chongqing, China ,Chonggang General Hospital, 400000 Chongqing, China
| | - Jane Yu
- grid.24827.3b0000 0001 2179 9593Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, OH 45267 USA
| | - Chenggang Li
- grid.216938.70000 0000 9878 7032State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Nankai University, 300350 Tianjin, P.R. China
| |
Collapse
|
5
|
Differential activation mechanisms of lipid GPCRs by lysophosphatidic acid and sphingosine 1-phosphate. Nat Commun 2022; 13:731. [PMID: 35136060 PMCID: PMC8826421 DOI: 10.1038/s41467-022-28417-2] [Citation(s) in RCA: 59] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Accepted: 01/21/2022] [Indexed: 12/23/2022] Open
Abstract
Lysophospholipids are bioactive lipids and can signal through G-protein-coupled receptors (GPCRs). The best studied lysophospholipids are lysophosphatidic acid (LPA) and sphingosine 1-phosphate (S1P). The mechanisms of lysophospholipid recognition by an active GPCR, and the activations of lysophospholipid GPCR–G-protein complexes remain unclear. Here we report single-particle cryo-EM structures of human S1P receptor 1 (S1P1) and heterotrimeric Gi complexes formed with bound S1P or the multiple sclerosis (MS) treatment drug Siponimod, as well as human LPA receptor 1 (LPA1) and Gi complexes in the presence of LPA. Our structural and functional data provide insights into how LPA and S1P adopt different conformations to interact with their cognate GPCRs, the selectivity of the homologous lipid GPCRs for S1P versus LPA, and the different activation mechanisms of these GPCRs by LPA and S1P. Our studies also reveal specific optimization strategies to improve the MS-treating S1P1-targeting drugs. Liu et al. report structures of human sphingosine 1-phosphate (S1P) receptor 1 (S1P1) in complex with Gi and S1P or the multiple sclerosis (MS) drug Siponimod, as well as human lysophosphatidic acid (LPA) receptor 1 (LPA1) in complex with Gi and LPA, revealing distinct conformations of the lysophospholipids interacting with their cognate GPCRs.
Collapse
|
6
|
Chen Z, Haider A, Chen J, Xiao Z, Gobbi L, Honer M, Grether U, Arnold SE, Josephson L, Liang SH. The Repertoire of Small-Molecule PET Probes for Neuroinflammation Imaging: Challenges and Opportunities beyond TSPO. J Med Chem 2021; 64:17656-17689. [PMID: 34905377 PMCID: PMC9094091 DOI: 10.1021/acs.jmedchem.1c01571] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Neuroinflammation is an adaptive response of the central nervous system to diverse potentially injurious stimuli, which is closely associated with neurodegeneration and typically characterized by activation of microglia and astrocytes. As a noninvasive and translational molecular imaging tool, positron emission tomography (PET) could provide a better understanding of neuroinflammation and its role in neurodegenerative diseases. Ligands to translator protein (TSPO), a putative marker of neuroinflammation, have been the most commonly studied in this context, but they suffer from serious limitations. Herein we present a repertoire of different structural chemotypes and novel PET ligand design for classical and emerging neuroinflammatory targets beyond TSPO. We believe that this Perspective will support multidisciplinary collaborations in academic and industrial institutions working on neuroinflammation and facilitate the progress of neuroinflammation PET probe development for clinical use.
Collapse
Affiliation(s)
- Zhen Chen
- Division of Nuclear Medicine and Molecular Imaging, Massachusetts General Hospital & Department of Radiology, Harvard Medical School, Boston, MA, 02114, United States
| | - Ahmed Haider
- Division of Nuclear Medicine and Molecular Imaging, Massachusetts General Hospital & Department of Radiology, Harvard Medical School, Boston, MA, 02114, United States
| | - Jiahui Chen
- Division of Nuclear Medicine and Molecular Imaging, Massachusetts General Hospital & Department of Radiology, Harvard Medical School, Boston, MA, 02114, United States
| | - Zhiwei Xiao
- Division of Nuclear Medicine and Molecular Imaging, Massachusetts General Hospital & Department of Radiology, Harvard Medical School, Boston, MA, 02114, United States
| | - Luca Gobbi
- Pharma Research and Early Development, F. Hoffmann-La Roche Ltd, CH-4070 Basel, Switzerland
| | - Michael Honer
- Pharma Research and Early Development, F. Hoffmann-La Roche Ltd, CH-4070 Basel, Switzerland
| | - Uwe Grether
- Pharma Research and Early Development, F. Hoffmann-La Roche Ltd, CH-4070 Basel, Switzerland
| | - Steven E. Arnold
- Department of Neurology and the Massachusetts Alzheimer’s Disease Research Center, Massachusetts General Hospital, Harvard Medical School, 114 16th Street, Charlestown, Massachusetts 02129, USA
| | - Lee Josephson
- Division of Nuclear Medicine and Molecular Imaging, Massachusetts General Hospital & Department of Radiology, Harvard Medical School, Boston, MA, 02114, United States
| | - Steven H. Liang
- Division of Nuclear Medicine and Molecular Imaging, Massachusetts General Hospital & Department of Radiology, Harvard Medical School, Boston, MA, 02114, United States
| |
Collapse
|
7
|
Lysophospholipids in Lung Inflammatory Diseases. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1303:373-391. [PMID: 33788203 DOI: 10.1007/978-3-030-63046-1_20] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
The lysophospholipids (LPLs) belong to a group of bioactive lipids that play pivotal roles in several physiological and pathological processes. LPLs are derivatives of phospholipids and consist of a single hydrophobic fatty acid chain, a hydrophilic head, and a phosphate group with or without a large molecule attached. Among the LPLs, lysophosphatidic acid (LPA) and sphingosine-1-phosphate (S1P) are the simplest, and have been shown to be involved in lung inflammatory symptoms and diseases such as acute lung injury, asthma, and chronic obstructive pulmonary diseases. G protein-coupled receptors (GPCRs) mediate LPA and S1P signaling. In this chapter, we will discuss on the role of LPA, S1P, their metabolizing enzymes, inhibitors or agonists of their receptors, and their GPCR-mediated signaling in lung inflammatory symptoms and diseases, focusing specially on acute respiratory distress syndrome, asthma, and chronic obstructive pulmonary disease.
Collapse
|
8
|
Selective detection of phospholipids using molecularly imprinted fluorescent sensory core-shell particles. Sci Rep 2020; 10:9924. [PMID: 32555511 PMCID: PMC7303128 DOI: 10.1038/s41598-020-66802-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Accepted: 05/28/2020] [Indexed: 12/28/2022] Open
Abstract
Sphingosine-1-phosphate (S1P) is a bioactive sphingo-lipid with a broad range of activities coupled to its role in G-protein coupled receptor signalling. Monitoring of both intra and extra cellular levels of this lipid is challenging due to its low abundance and lack of robust affinity assays or sensors. We here report on fluorescent sensory core-shell molecularly imprinted polymer (MIP) particles responsive to near physiologically relevant levels of S1P and the S1P receptor modulator fingolimod phosphate (FP) in spiked human serum samples. Imprinting was achieved using the tetrabutylammonium (TBA) salt of FP or phosphatidic acid (DPPA·Na) as templates in combination with a polymerizable nitrobenzoxadiazole (NBD)-urea monomer with the dual role of capturing the phospho-anion and signalling its presence. The monomers were grafted from ca 300 nm RAFT-modified silica core particles using ethyleneglycol dimethacrylate (EGDMA) as crosslinker resulting in 10–20 nm thick shells displaying selective fluorescence response to the targeted lipids S1P and DPPA in aqueous buffered media. Potential use of the sensory particles for monitoring S1P in serum was demonstrated on spiked serum samples, proving a linear range of 18–60 µM and a detection limit of 5.6 µM, a value in the same range as the plasma concentration of the biomarker.
Collapse
|
9
|
Fingolimod (FTY720) improves the functional recovery and myelin preservation of the optic pathway in focal demyelination model of rat optic chiasm. Brain Res Bull 2019; 153:109-121. [DOI: 10.1016/j.brainresbull.2019.08.014] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Revised: 08/09/2019] [Accepted: 08/18/2019] [Indexed: 12/21/2022]
|
10
|
Rafiee Zadeh A, Parsa S, Tavoosi N, Farshi M, Masaeli MF. Effect of fingolimod on white blood cell, lymphocyte and neutrophil counts in MS patients. AMERICAN JOURNAL OF CLINICAL AND EXPERIMENTAL IMMUNOLOGY 2019; 8:9-15. [PMID: 31131156 PMCID: PMC6526352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Accepted: 03/15/2019] [Indexed: 06/09/2023]
Abstract
INTRODUCTION Fingolimod is an immunomodulating oral treatment used for treating relapsing-remitting multiple sclerosis (RRMS). The exact mechanism for its action in preventing relapses is unknown. Also, its affect on immune cell populations remains unestablished. OBJECTIVES This study will measure the changes in cell populations of WBCs, lymphocytes, and neutrophils in MS patients after one month of treatment. METHODS 66 MS patients from Isfahan Province with RRMS were chosen based on certain exclusion criteria and eligibility for fingolimod oral treatment. Initial cell counts for WBC, lymphocyte, and neutrophil cell populations were achieved. Fingolimod .5 mg daily treatment was then initiated under the supervision of a physician. After one month of treatment, cell counts were repeated. Statistical analysis was performed using SPSS. RESULTS Both lymphocyte and WBC mean cell counts were significantly decreased in this patient cohort. Neutrophil average cell counts were significantly increased in this 66 patient cohort. Only the decrease of WBC populations was significant for both male and female cohorts individually. Only female sub-cohorts were significantly changed for neutrophils and lymphocytes, increased and decreased respectively. Male sub-cohorts maintained the same directionality but failed to produce statistical significance. CONCLUSION While fingolimod has been effectively proven as reducing lymphocyte cells in most patient populations, its effects on neutrophils have not been studied in abundance. Also, there may be sex-related differences in responses to fingolimod treatment with regards to lymphocytes and neutrophils, suggesting a possible difference in RRMS pathogenesis between males and females.
Collapse
Affiliation(s)
| | - Sara Parsa
- School of Medicine, Islamic Azad University, Najafabad BranchIsfahan, Iran
| | - Nooshin Tavoosi
- Department of Midwifery, School of Nursing and Midwifery, Shahrekord University of Medical SciencesShahrekord, Iran
| | - Mohsen Farshi
- School of Medicine, Isfahan University of Medical SciencesIsfahan, Iran
| | | |
Collapse
|
11
|
Ahmed N, Linardi D, Muhammad N, Chiamulera C, Fumagalli G, Biagio LS, Gebrie MA, Aslam M, Luciani GB, Faggian G, Rungatscher A. Sphingosine 1-Phosphate Receptor Modulator Fingolimod (FTY720) Attenuates Myocardial Fibrosis in Post-heterotopic Heart Transplantation. Front Pharmacol 2017; 8:645. [PMID: 28966593 PMCID: PMC5605636 DOI: 10.3389/fphar.2017.00645] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2017] [Accepted: 08/31/2017] [Indexed: 12/21/2022] Open
Abstract
Background and Objective: Sphingosine 1-phosphate (S1P), and S1P receptor modulator fingolimod have been suggested to play important cardioprotective role in animal models of myocardial ischemia/reperfusion injuries. To understand the cardioprotective function of S1P and its mechanism in vivo, we analyzed apoptotic, inflammatory biomarkers, and myocardial fibrosis in an in vivo heterotopic rat heart transplantation model. Methods: Heterotopic heart transplantation is performed in 60 Sprague–Dawley (SD) rats (350–400 g). The heart transplant recipients (n = 60) are categorized into Group A (control) and Group B (fingolimod treated 1 mg/kg intravenous). At baseline with 24 h after heart transplantation, blood and myocardial tissue are collected for analysis of myocardial biomarkers, apoptosis, inflammatory markers, oxidative stress, and phosphorylation of Akt/Erk/STAT-3 signaling pathways. Myocardial fibrosis was investigated using Masson’s trichrome staining and L-hydroxyline. Results: Fingolimod treatment activates both Reperfusion Injury Salvage Kinase (RISK) and Survivor Activating Factor Enhancement (SAFE) pathways as evident from activation of anti-apoptotic and anti-inflammatory pathways. Fingolimod treatment caused a reduction in myocardial oxidative stress and hence cardiomyocyte apoptosis resulting in a decrease in myocardial reperfusion injury. Moreover, a significant (p < 0.001) reduction in collagen staining and hydroxyproline content was observed in fingolimod treated animals 30 days after transplantation demonstrating a reduction in cardiac fibrosis. Conclusion: S1P receptor activation with fingolimod activates anti-apoptotic and anti-inflammatory pathways, leading to improved myocardial salvage causing a reduction in cardiac fibrosis.
Collapse
Affiliation(s)
- Naseer Ahmed
- Section of Cardiac Surgery, Department of Surgery, Dentistry, Paediatrics and Gynaecology, University of VeronaVerona, Italy.,Faculty of Health Sciences, University of PunjabLahore, Pakistan.,Research Unit, Faculty of Allied Health Sciences, University of LahoreLahore, Pakistan.,Section of Pharmacology, Department of Diagnostics and Public Health, University of VeronaVerona, Italy
| | - Daniele Linardi
- Section of Cardiac Surgery, Department of Surgery, Dentistry, Paediatrics and Gynaecology, University of VeronaVerona, Italy
| | - Nazeer Muhammad
- COMSATS Institute of Information TechnologyWah Cantt, Pakistan
| | - Cristiano Chiamulera
- Section of Pharmacology, Department of Diagnostics and Public Health, University of VeronaVerona, Italy
| | - Guido Fumagalli
- Section of Pharmacology, Department of Diagnostics and Public Health, University of VeronaVerona, Italy
| | - Livio San Biagio
- Section of Cardiac Surgery, Department of Surgery, Dentistry, Paediatrics and Gynaecology, University of VeronaVerona, Italy
| | - Mebratu A Gebrie
- Section of Cardiac Surgery, Department of Surgery, Dentistry, Paediatrics and Gynaecology, University of VeronaVerona, Italy.,Department of Anatomy, Università di Addis AbebaAddis Ababa, Ethiopia
| | - Muhammad Aslam
- Department of Internal Medicine, Cardiology and Angiology, University Hospital, Justus Liebig UniversityGiessen, Germany
| | - Giovanni Battista Luciani
- Section of Cardiac Surgery, Department of Surgery, Dentistry, Paediatrics and Gynaecology, University of VeronaVerona, Italy
| | - Giuseppe Faggian
- Section of Cardiac Surgery, Department of Surgery, Dentistry, Paediatrics and Gynaecology, University of VeronaVerona, Italy
| | - Alessio Rungatscher
- Section of Cardiac Surgery, Department of Surgery, Dentistry, Paediatrics and Gynaecology, University of VeronaVerona, Italy
| |
Collapse
|
12
|
Mao-Draayer Y, Sarazin J, Fox D, Schiopu E. The sphingosine-1-phosphate receptor: A novel therapeutic target for multiple sclerosis and other autoimmune diseases. Clin Immunol 2016; 175:10-15. [PMID: 27890706 DOI: 10.1016/j.clim.2016.11.008] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2016] [Revised: 11/12/2016] [Accepted: 11/18/2016] [Indexed: 12/30/2022]
Abstract
Multiple sclerosis (MS) is a prototype autoimmune disease of the central nervous system (CNS). Currently, there is no drug that provides a cure for MS. To date, all immunotherapeutic drugs target relapsing remitting MS (RR-MS); it remains a daunting medical challenge in MS to develop therapy for secondary progressive MS (SP-MS). Since the approval of the non-selective sphingosine-1-phosphate (S1P) receptor modulator FTY720 (fingolimod [Gilenya®]) for RR-MS in 2010, there have been many emerging studies with various selective S1P receptor modulators in other autoimmune conditions. In this article, we will review how S1P receptor may be a promising therapeutic target for SP-MS and other autoimmune diseases such as psoriasis, polymyositis and lupus.
Collapse
Affiliation(s)
- Yang Mao-Draayer
- Department of Neurology and Clinical Autoimmunity Center of Excellence, University of Michigan Medical School, 4015 Alfred Taubman Biomedical Sciences Research Bldg., 109 Zina Pitcher Place, Ann Arbor, MI 48109-2200, United States.
| | - Jeffrey Sarazin
- Department of Neurology and Clinical Autoimmunity Center of Excellence, University of Michigan Medical School, 4015 Alfred Taubman Biomedical Sciences Research Bldg., 109 Zina Pitcher Place, Ann Arbor, MI 48109-2200, United States
| | - David Fox
- Department of Neurology and Clinical Autoimmunity Center of Excellence, University of Michigan Medical School, 4015 Alfred Taubman Biomedical Sciences Research Bldg., 109 Zina Pitcher Place, Ann Arbor, MI 48109-2200, United States
| | - Elena Schiopu
- Department of Neurology and Clinical Autoimmunity Center of Excellence, University of Michigan Medical School, 4015 Alfred Taubman Biomedical Sciences Research Bldg., 109 Zina Pitcher Place, Ann Arbor, MI 48109-2200, United States
| |
Collapse
|
13
|
Samuvel DJ, Saxena N, Dhindsa JS, Singh AK, Gill GS, Grobelny DW, Singh I. AKP-11 - A Novel S1P1 Agonist with Favorable Safety Profile Attenuates Experimental Autoimmune Encephalomyelitis in Rat Model of Multiple Sclerosis. PLoS One 2015; 10:e0141781. [PMID: 26513477 PMCID: PMC4626178 DOI: 10.1371/journal.pone.0141781] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2015] [Accepted: 10/13/2015] [Indexed: 12/22/2022] Open
Abstract
Sphingosine-1-phosphate receptor 1 (S1P1) mediated regulation of lymphocyte egress from lymphoid organs is recognized as the mechanism of FTY720 (Fingolimod, Gilenya) efficacy in relapsing-remitting forms of multiple sclerosis (RRMS). In this study we describe a novel S1P1 agonist AKP-11, next generation of S1P1 agonist, with immunomodulatory activities in cell culture model and for therapeutic efficacy against an animal model of MS, i.e. experimental autoimmune encephalomyelitis (EAE) but without the adverse effects observed with FTY720. Like FTY720, AKP-11 bound to S1P1 is internalized and activates intracellular AKT and ERKs cellular signaling pathways. In contrast to FTY720, AKP-11 mediated S1P1 downregulation is independent of sphingosine kinase activity indicating it to be a direct agonist of S1P1. The S1P1 loss and inhibition of lymphocyte egress by FTY720 leads to lymphopenia. In comparison with FTY720, oral administration of AKP-11 caused milder and reversible lymphopenia while providing a similar degree of therapeutic efficacy in the EAE animal model. Consistent with the observed reversible lymphopenia with AKP-11, the S1P1 recycled back to cell membrane in AKP-11 treated cells following its withdrawal, but not with withdrawal of FTY720. Accordingly, a smaller degree of ubiquitination and proteolysis of S1P1 was observed in AKP-11 treated cells as compared to FTY720. Consistent with previous observations, FTY720 treatment is associated with adverse effects of bradycardia and lung vascular leaks in rodents, whereas AKP-11 treatment had undetectable effects on bradycardia and reduced lung vascular leaks as compared to FTY720. Taken together, the data documents that AKP-11 treatment cause milder and reversible lymphopenia with milder adverse effects while maintaining therapeutic efficacy similar to that observed with FTY720, thus indicating therapeutic potential of AKP-11 for treatment of MS and related autoimmune disorders.
Collapse
Affiliation(s)
- Devadoss J. Samuvel
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, South Carolina, United States of America
| | - Nishant Saxena
- Charles P. Darby Children’s Research Institute, Department of Pediatrics, Medical University of South Carolina, Charleston, South Carolina, United States of America
| | - Jasdeep S. Dhindsa
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, South Carolina, United States of America
| | - Avtar K. Singh
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, South Carolina, United States of America
| | - Gurmit S. Gill
- Akaal Pharma Pty Ltd., 310E Thomas Cherry Building, Bundoora, Australia
| | | | - Inderjit Singh
- Charles P. Darby Children’s Research Institute, Department of Pediatrics, Medical University of South Carolina, Charleston, South Carolina, United States of America
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, South Carolina, United States of America
- * E-mail:
| |
Collapse
|
14
|
Subei AM, Ontaneda D. Risk Mitigation Strategies for Adverse Reactions Associated with the Disease-Modifying Drugs in Multiple Sclerosis. CNS Drugs 2015; 29:759-71. [PMID: 26407624 PMCID: PMC4621807 DOI: 10.1007/s40263-015-0277-4] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Over the past several years, the number of disease-modifying therapies (DMTs) for the treatment of multiple sclerosis (MS) has doubled in number. The 13 approved agents have shown a wide range of efficacy and safety in their clinical trials and post-marketing experience. While the availability of the newer agents allows for a wider selection of therapy for clinicians and patients, there is a need for careful understanding of the benefits and risks of each agent. Several factors such as the medication efficacy, side-effect profile, patient's preference, and co-morbidities need to be considered. An individualized treatment approach is thus imperative. In this review, risk stratification and mitigation strategies of the various disease-modifying agents are discussed.
Collapse
Affiliation(s)
- Adnan M Subei
- Mellen Center for MS Treatment and Research, Cleveland Clinic Foundation, 9500 Euclid Avenue/U10, Cleveland, OH, 44195, USA.
| | - Daniel Ontaneda
- Mellen Center for MS Treatment and Research, Cleveland Clinic Foundation, 9500 Euclid Avenue/U10, Cleveland, OH, 44195, USA.
| |
Collapse
|
15
|
Attiori Essis S, Laurier-Laurin ME, Pépin É, Cyr M, Massicotte G. GluN2B-containing NMDA receptors are upregulated in plasma membranes by the sphingosine-1-phosphate analog FTY720P. Brain Res 2015; 1624:349-358. [PMID: 26260438 DOI: 10.1016/j.brainres.2015.07.055] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2015] [Revised: 07/28/2015] [Accepted: 07/30/2015] [Indexed: 11/29/2022]
Abstract
Sphingosine-1-phosphate (S1P) is a ceramide derivative serving not only as a regulator of immune properties but also as a modulator of brain functions. To better understand the mechanism underlying the effects of S1P on brain functions, we investigated the potential impact of S1P receptor (S1PR) activation on NMDA receptor subunits. We used acute rat hippocampal slices as a model system, and determined the effects of the active phosphorylated S1P analog, fingolimod (FTY720P) on various NMDA receptors. Treatment with FTY720P significantly increased phosphorylation of GluN2B-containing NMDA receptors at Tyr1472. This effect appears rather specific, as treatment with FTY720P did not modify GluN2B-Tyr1336, GluN2B-Ser1480, GluN2A-Tyr1325 or GluN1-Ser897 phosphorylation. Pre-treatment of hippocampal slices with the compounds W146 and PP1 indicated that FTY720P-induced GluN2B phosphorylation at Tyr1472 epitopes was dependent on activation of S1PR subunit 1 (S1PR1) and Src/Fyn kinase, respectively. Cell surface biotinylation experiments indicated that FTY720P-induced GluN2B phosphorylation at Tyr1472 was also associated with increased levels of GluN1 and GluN2B subunits on membrane surface, whereas no change was observed for GluN2A subunits. We finally demonstrate that FTY720P is inclined to favor Tau and Fyn accumulation on plasma membranes. These results suggest that activation of S1PR1 by FTY720P enhances GluN2B receptor phosphorylation in rat hippocampal slices, resulting in increased levels of GluN1 and GluN2B receptor subunits in neuronal membranes through a mechanism probably involving Fyn and Tau.
Collapse
Affiliation(s)
- Suzanne Attiori Essis
- Département de biologie médicale, Université du Québec à Trois-Rivières, Trois-Rivières, Québec, Canada G9A 5H7
| | - Marie-Elaine Laurier-Laurin
- Département de biologie médicale, Université du Québec à Trois-Rivières, Trois-Rivières, Québec, Canada G9A 5H7
| | - Élise Pépin
- Département de biologie médicale, Université du Québec à Trois-Rivières, Trois-Rivières, Québec, Canada G9A 5H7
| | - Michel Cyr
- Département de biologie médicale, Université du Québec à Trois-Rivières, Trois-Rivières, Québec, Canada G9A 5H7
| | - Guy Massicotte
- Département de biologie médicale, Université du Québec à Trois-Rivières, Trois-Rivières, Québec, Canada G9A 5H7.
| |
Collapse
|
16
|
Abstract
Sphingosine 1-phosphate (S1P) receptor modulators possess a unique mechanism of action as disease-modifying therapy for multiple sclerosis (MS). Subtype 1 S1P receptors are expressed on the surfaces of lymphocytes and are important in regulating egression from lymph nodes. The S1P receptor modulators indirectly antagonize the receptor's function and sequester lymphocytes in lymph nodes. Fingolimod was the first S1P agent approved in the USA in 2010 for relapsing MS after two phase III trials (FREEDOMS and TRANSFORMS) demonstrated potent efficacy, and good safety and tolerability. Post-marketing experience, as well as a third phase III trial (FREEDOMS II), also showed favorable results. More selective S1P receptor agents-ponesimod (ACT128800), siponimod (BAF312), ozanimod (RPC1063), ceralifimod (ONO-4641), GSK2018682, and MT-1303-are still in relatively early stages of development, but phase I and II trials showed promising efficacy and safety. However, these observations have yet to be reproduced in phase III clinical trials.
Collapse
Affiliation(s)
- Adnan M. Subei
- Mellen Center for MS Treatment and Research, Cleveland Clinic Foundation, 9500 Euclid Avenue/U10, Cleveland, OH 44195, Phone: 216-445-8110, Fax: 216-445-7013
| | - Jeffrey A. Cohen
- Mellen Center for MS Treatment and Research, Cleveland Clinic Foundation, 9500 Euclid Avenue/U10, Cleveland, OH 44195, Phone: 216-445-8110, Fax: 216-445-7013
| |
Collapse
|
17
|
Ikeuchi K, Hayashi M, Yamamoto T, Inai M, Asakawa T, Hamashima Y, Kan T. Stereocontrolled Total Synthesis of Sphingofungin E. European J Org Chem 2013. [DOI: 10.1002/ejoc.201301065] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
18
|
O'Sullivan C, Dev KK. The structure and function of the S1P1 receptor. Trends Pharmacol Sci 2013; 34:401-12. [PMID: 23763867 DOI: 10.1016/j.tips.2013.05.002] [Citation(s) in RCA: 106] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2013] [Revised: 04/27/2013] [Accepted: 05/07/2013] [Indexed: 12/18/2022]
Abstract
Sphingosine 1-phosphate (S1P) receptors (S1PRs) belong to the class A family of G protein-coupled receptors (GPCRs). S1PRs are widely expressed on many cell types, including those of the immune, cardiovascular, and central nervous systems. The S1PR family is rapidly gaining attention as an important mediator of many cellular processes, including cell differentiation, migration, survival, angiogenesis, calcium homeostasis, inflammation and immunity. Importantly, S1PRs are known drug targets for multiple sclerosis (MS), for which the newly developed oral therapy fingolimod, an S1PR modulator, has recently been approved for clinical use. Much progress has also recently been made in the field of structural biology and in the modeling of heterotrimeric GPCRs allowing the crystal structure of the S1PR1 subtype to be elucidated and key interactions defined. Here, we outline the structure and function of S1PR1, highlighting the key residues involved in receptor activation, signaling, transmembrane interactions, ligand binding, post-translational modification, and protein-protein interactions.
Collapse
Affiliation(s)
- Catherine O'Sullivan
- Molecular Neuropharmacology, Department of Physiology, School of Medicine, Trinity College Institute of Neuroscience, Trinity College Dublin, Dublin, Ireland
| | | |
Collapse
|