1
|
Danielson SM, Lefferts AR, Norman E, Regner EH, Schulz HM, Sansone-Poe D, Orlicky DJ, Kuhn KA. Myeloid Cells and Sphingosine-1-Phosphate Are Required for TCRαβ Intraepithelial Lymphocyte Recruitment to the Colon Epithelium. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 212:1843-1854. [PMID: 38568091 PMCID: PMC11105980 DOI: 10.4049/jimmunol.2200556] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Accepted: 03/16/2024] [Indexed: 04/07/2024]
Abstract
Intraepithelial lymphocytes (IELs) are T cells important for the maintenance of barrier integrity in the intestine. Colon IELs are significantly reduced in both MyD88-deficient mice and those lacking an intact microbiota, suggesting that MyD88-mediated detection of bacterial products is important for the recruitment and/or retention of these cells. Here, using conditionally deficient MyD88 mice, we show that myeloid cells are the key mediators of TCRαβ+ IEL recruitment to the colon. Upon exposure to luminal bacteria, myeloid cells produce sphingosine-1-phosphate (S1P) in a MyD88-dependent fashion. TCRαβ+ IEL recruitment may be blocked using the S1P receptor antagonist FTY720, confirming the importance of S1P in the recruitment of TCRαβ+ IELs to the colon epithelium. Finally, using the TNFΔARE/+ model of Crohn's-like bowel inflammation, we show that disruption of colon IEL recruitment through myeloid-specific MyD88 deficiency results in reduced pathology. Our results illustrate one mechanism for recruitment of a subset of IELs to the colon.
Collapse
Affiliation(s)
- Sarah Mann Danielson
- Division of Rheumatology, Department of Medicine University of Colorado Anschutz Medical Campus, Aurora, CO
| | - Adam R. Lefferts
- Division of Rheumatology, Department of Medicine University of Colorado Anschutz Medical Campus, Aurora, CO
| | - Eric Norman
- Division of Rheumatology, Department of Medicine University of Colorado Anschutz Medical Campus, Aurora, CO
| | - Emilie H. Regner
- Division of Rheumatology, Department of Medicine University of Colorado Anschutz Medical Campus, Aurora, CO
- Division of Gastroenterology and Hepatology, Department of Medicine University of Colorado Anschutz Medical Campus, Aurora, CO
- Current affiliation: Division of Gastroenterology, Department of Medicine, Oregon Health Sciences University, Portland, OR
| | - Hanna M. Schulz
- Division of Rheumatology, Department of Medicine University of Colorado Anschutz Medical Campus, Aurora, CO
| | - Danielle Sansone-Poe
- Division of Rheumatology, Department of Medicine University of Colorado Anschutz Medical Campus, Aurora, CO
| | - David J. Orlicky
- Department of Pathology, University of Colorado School of Medicine, Aurora, CO
| | - Kristine A. Kuhn
- Division of Rheumatology, Department of Medicine University of Colorado Anschutz Medical Campus, Aurora, CO
| |
Collapse
|
2
|
Magalhães DM, Stewart NA, Mampay M, Rolle SO, Hall CM, Moeendarbary E, Flint MS, Sebastião AM, Valente CA, Dymond MK, Sheridan GK. The sphingosine 1-phosphate analogue, FTY720, modulates the lipidomic signature of the mouse hippocampus. J Neurochem 2024; 168:1113-1142. [PMID: 38339785 DOI: 10.1111/jnc.16073] [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: 08/17/2023] [Revised: 12/27/2023] [Accepted: 01/22/2024] [Indexed: 02/12/2024]
Abstract
The small-molecule drug, FTY720 (fingolimod), is a synthetic sphingosine 1-phosphate (S1P) analogue currently used to treat relapsing-remitting multiple sclerosis in both adults and children. FTY720 can cross the blood-brain barrier (BBB) and, over time, accumulate in lipid-rich areas of the central nervous system (CNS) by incorporating into phospholipid membranes. FTY720 has been shown to enhance cell membrane fluidity, which can modulate the functions of glial cells and neuronal populations involved in regulating behaviour. Moreover, direct modulation of S1P receptor-mediated lipid signalling by FTY720 can impact homeostatic CNS physiology, including neurotransmitter release probability, the biophysical properties of synaptic membranes, ion channel and transmembrane receptor kinetics, and synaptic plasticity mechanisms. The aim of this study was to investigate how chronic FTY720 treatment alters the lipid composition of CNS tissue in adolescent mice at a key stage of brain maturation. We focused on the hippocampus, a brain region known to be important for learning, memory, and the processing of sensory and emotional stimuli. Using mass spectrometry-based lipidomics, we discovered that FTY720 increases the fatty acid chain length of hydroxy-phosphatidylcholine (PCOH) lipids in the mouse hippocampus. It also decreases PCOH monounsaturated fatty acids (MUFAs) and increases PCOH polyunsaturated fatty acids (PUFAs). A total of 99 lipid species were up-regulated in the mouse hippocampus following 3 weeks of oral FTY720 exposure, whereas only 3 lipid species were down-regulated. FTY720 also modulated anxiety-like behaviours in young mice but did not affect spatial learning or memory formation. Our study presents a comprehensive overview of the lipid classes and lipid species that are altered in the hippocampus following chronic FTY720 exposure and provides novel insight into cellular and molecular mechanisms that may underlie the therapeutic or adverse effects of FTY720 in the central nervous system.
Collapse
Affiliation(s)
- Daniela M Magalhães
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
- Instituto de Medicina Molecular João Lobo Antunes, Lisboa, Portugal
- School of Applied Sciences, University of Brighton, Brighton, UK
| | | | - Myrthe Mampay
- School of Applied Sciences, University of Brighton, Brighton, UK
| | - Sara O Rolle
- Green Templeton College, University of Oxford, Oxford, UK
| | - Chloe M Hall
- School of Applied Sciences, University of Brighton, Brighton, UK
- Department of Mechanical Engineering, University College London, London, UK
| | - Emad Moeendarbary
- Department of Mechanical Engineering, University College London, London, UK
- 199 Biotechnologies Ltd, London, UK
| | - Melanie S Flint
- School of Applied Sciences, University of Brighton, Brighton, UK
| | - Ana M Sebastião
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
- Instituto de Medicina Molecular João Lobo Antunes, Lisboa, Portugal
| | - Cláudia A Valente
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
- Instituto de Medicina Molecular João Lobo Antunes, Lisboa, Portugal
| | - Marcus K Dymond
- School of Applied Sciences, University of Brighton, Brighton, UK
| | | |
Collapse
|
3
|
Lee J, Lester R, O'Reilly T, Lowe ER, Slatkin NE, Franklin H, Israel RJ. A Randomized, Double-Blind, Parallel Design Thorough QT Study With a Nested Crossover to Compare the Cardiac Safety of Amiselimod With Placebo and Positive Control in Healthy Volunteers. Clin Pharmacol Drug Dev 2023; 12:236-248. [PMID: 36708138 DOI: 10.1002/cpdd.1210] [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: 03/03/2022] [Accepted: 11/28/2022] [Indexed: 01/29/2023]
Abstract
This double-blind study evaluated the cardiac safety of amiselimod. Healthy adults (n = 190) were randomized (2:1:1) to receive (1) oral placebo (day -1), followed by oral amiselimod (days 1-26), which was upwardly titrated from 0.4 to 1.6 mg once daily to achieve steady-state concentrations comparable with 0.4 (therapeutic) and 0.8 mg (supratherapeutic) once daily, and placebo (day 27); (2) placebo (day -1), oral moxifloxacin 400 mg (day 1; positive control), followed by placebo (days 1-27); or (3) placebo (days -1 to 26), followed by moxifloxacin 400 mg (day 27). No participant had a corrected QT interval by Fredericia (QTcF) >500 milliseconds or a change from baseline (dQTcF) >60 milliseconds. The upper limits of the 90%CIs for the differences in least-squares mean difference in dQTcF between amiselimod and placebo on days 13 and 26 were <10 milliseconds. Area under the concentration-time curve from 0 to 23.5 hours after dosing and maximum plasma concentration of amiselimod and amiselimod-P (active metabolite) at steady-state concentrations for the 0.8-mg dose on day 26 were approximately double that observed with the 0.4-mg dose on day 13. All adverse events were mild to moderate in severity, and no deaths occurred. Amiselimod did not have any clinically relevant effect on the QTcF interval.
Collapse
Affiliation(s)
- Jimin Lee
- Bausch Health US, LLC, Bridgewater, New Jersey, USA
| | | | | | - Ezra R Lowe
- Bausch Health US, LLC, Bridgewater, New Jersey, USA
| | | | | | | |
Collapse
|
4
|
Cavalcanti F, Gonzalez-Rey E, Delgado M, Falo CP, Mestre L, Guaza C, O’Valle F, Lufino MMP, Xaus J, Mascaró C, Lunardi S, Sacilotto N, Dessanti P, Rotllant D, Navarro X, Herrando-Grabulosa M, Buesa C, Maes T. Efficacy of Vafidemstat in Experimental Autoimmune Encephalomyelitis Highlights the KDM1A/RCOR1/HDAC Epigenetic Axis in Multiple Sclerosis. Pharmaceutics 2022; 14:pharmaceutics14071420. [PMID: 35890315 PMCID: PMC9323733 DOI: 10.3390/pharmaceutics14071420] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Revised: 06/27/2022] [Accepted: 06/27/2022] [Indexed: 02/01/2023] Open
Abstract
Lysine specific demethylase 1 (LSD1; also known as KDM1A), is an epigenetic modulator that modifies the histone methylation status. KDM1A forms a part of protein complexes that regulate the expression of genes involved in the onset and progression of diseases such as cancer, central nervous system (CNS) disorders, viral infections, and others. Vafidemstat (ORY-2001) is a clinical stage inhibitor of KDM1A in development for the treatment of neurodegenerative and psychiatric diseases. However, the role of ORY-2001 targeting KDM1A in neuroinflammation remains to be explored. Here, we investigated the effect of ORY-2001 on immune-mediated and virus-induced encephalomyelitis, two experimental models of multiple sclerosis and neuronal damage. Oral administration of ORY-2001 ameliorated clinical signs, reduced lymphocyte egress and infiltration of immune cells into the spinal cord, and prevented demyelination. Interestingly, ORY-2001 was more effective and/or faster acting than a sphingosine 1-phosphate receptor antagonist in the effector phase of the disease and reduced the inflammatory gene expression signature characteristic ofEAE in the CNS of mice more potently. In addition, ORY-2001 induced gene expression changes concordant with a potential neuroprotective function in the brain and spinal cord and reduced neuronal glutamate excitotoxicity-derived damage in explants. These results pointed to ORY-2001 as a promising CNS epigenetic drug able to target neuroinflammatory and neurodegenerative diseases and provided preclinical support for the subsequent design of early-stage clinical trials.
Collapse
Affiliation(s)
- Fernando Cavalcanti
- Oryzon Genomics S.A., Carrer Sant Ferran 74, 08940 Cornellà de Llobregat, Spain; (F.C.); (M.M.P.L.); (J.X.); (C.M.); (N.S.); (P.D.); (D.R.); (C.B.)
| | - Elena Gonzalez-Rey
- Institute of Parasitology and Biomedicine López-Neyra, IPBLN-CSIC, PTS-Granada, 18016 Granada, Spain; (E.G.-R.); (M.D.)
| | - Mario Delgado
- Institute of Parasitology and Biomedicine López-Neyra, IPBLN-CSIC, PTS-Granada, 18016 Granada, Spain; (E.G.-R.); (M.D.)
| | - Clara P. Falo
- Institute of Parasitology and Biomedicine López-Neyra, IPBLN-CSIC, PTS-Granada, 18016 Granada, Spain; (E.G.-R.); (M.D.)
| | - Leyre Mestre
- Department of Functional and Systems Neurobiology, Cajal Institute (CSIC), 28034 Madrid, Spain; (L.M.); (C.G.)
| | - Carmen Guaza
- Department of Functional and Systems Neurobiology, Cajal Institute (CSIC), 28034 Madrid, Spain; (L.M.); (C.G.)
| | - Francisco O’Valle
- Department of Pathology, School of Medicine, IBIMER and IBS-Granada, Granada University, 18071 Granada, Spain;
| | - Michele M. P. Lufino
- Oryzon Genomics S.A., Carrer Sant Ferran 74, 08940 Cornellà de Llobregat, Spain; (F.C.); (M.M.P.L.); (J.X.); (C.M.); (N.S.); (P.D.); (D.R.); (C.B.)
| | - Jordi Xaus
- Oryzon Genomics S.A., Carrer Sant Ferran 74, 08940 Cornellà de Llobregat, Spain; (F.C.); (M.M.P.L.); (J.X.); (C.M.); (N.S.); (P.D.); (D.R.); (C.B.)
| | - Cristina Mascaró
- Oryzon Genomics S.A., Carrer Sant Ferran 74, 08940 Cornellà de Llobregat, Spain; (F.C.); (M.M.P.L.); (J.X.); (C.M.); (N.S.); (P.D.); (D.R.); (C.B.)
| | - Serena Lunardi
- Oryzon Genomics S.A., Carrer Sant Ferran 74, 08940 Cornellà de Llobregat, Spain; (F.C.); (M.M.P.L.); (J.X.); (C.M.); (N.S.); (P.D.); (D.R.); (C.B.)
| | - Natalia Sacilotto
- Oryzon Genomics S.A., Carrer Sant Ferran 74, 08940 Cornellà de Llobregat, Spain; (F.C.); (M.M.P.L.); (J.X.); (C.M.); (N.S.); (P.D.); (D.R.); (C.B.)
| | - Paola Dessanti
- Oryzon Genomics S.A., Carrer Sant Ferran 74, 08940 Cornellà de Llobregat, Spain; (F.C.); (M.M.P.L.); (J.X.); (C.M.); (N.S.); (P.D.); (D.R.); (C.B.)
| | - David Rotllant
- Oryzon Genomics S.A., Carrer Sant Ferran 74, 08940 Cornellà de Llobregat, Spain; (F.C.); (M.M.P.L.); (J.X.); (C.M.); (N.S.); (P.D.); (D.R.); (C.B.)
| | - Xavier Navarro
- Departament de Biologia Cellular, Fisiologia i Immunologia, Institut de Neurociències, Universitat Autònoma de Barcelona, and Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), 08193 Barcelona, Spain; (X.N.); (M.H.-G.)
| | - Mireia Herrando-Grabulosa
- Departament de Biologia Cellular, Fisiologia i Immunologia, Institut de Neurociències, Universitat Autònoma de Barcelona, and Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), 08193 Barcelona, Spain; (X.N.); (M.H.-G.)
| | - Carlos Buesa
- Oryzon Genomics S.A., Carrer Sant Ferran 74, 08940 Cornellà de Llobregat, Spain; (F.C.); (M.M.P.L.); (J.X.); (C.M.); (N.S.); (P.D.); (D.R.); (C.B.)
| | - Tamara Maes
- Oryzon Genomics S.A., Carrer Sant Ferran 74, 08940 Cornellà de Llobregat, Spain; (F.C.); (M.M.P.L.); (J.X.); (C.M.); (N.S.); (P.D.); (D.R.); (C.B.)
- Correspondence:
| |
Collapse
|
5
|
Suzuki S, Ogawa M, Miyazaki M, Ota K, Kazama H, Hirota A, Takano N, Hiramoto M, Miyazawa K. Lysosome‑targeted drug combination induces multiple organelle dysfunctions and non‑canonical death in pancreatic cancer cells. Oncol Rep 2021; 47:40. [PMID: 34958115 PMCID: PMC8759104 DOI: 10.3892/or.2021.8251] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Accepted: 12/03/2021] [Indexed: 11/09/2022] Open
Abstract
Pancreatic cancer is one of the leading causes of cancer-related mortality and has the lowest 5-year survival rate. Therefore, novel strategies are urgently required to treat pancreatic cancer. Pancreatic ductal adenocarcinoma (PDAC) cells rely on enhanced lysosomal function for survival and proliferation to facilitate the degradation of contents accumulated via autophagy and macropinocytosis. Previously, we have reported that the combination of epidermal growth factor receptor/HER2 inhibitor lapatinib and sphingosine analog fingolimod (FTY720) confers a significant cytostatic effect in lung cancer cells. In the present study, the combined effects of these drugs on PDAC cell lines, BxPC-3, KP-4, PANC-1 and MIA PaCa-2, were examined. It was observed that FTY720 enhanced the lapatinib-induced cytotoxic effect and caused non-canonical and lysosome-dependent death in PDAC cells. Lapatinib and FTY720 induced lysosomal swelling and inhibited lysosomal acidification. Combination treatment with lapatinib and FTY720 increased lysosomal membrane permeability, induced mitochondrial depolarization, induced endoplasmic reticulum stress and disturbed intracellular calcium homeostasis. Additionally, the cytotoxic effect of lapatinib was enhanced by hydroxychloroquine or the CDK4/6 inhibitor abemaciclib, both of which induce lysosomal dysfunction. Collectively, these results indicated that the lysosome-targeted drug combination induces multiple organelle dysfunction and exerts a marked cytotoxic effect in PDAC cells.
Collapse
Affiliation(s)
- Sumire Suzuki
- Department of Biochemistry, Tokyo Medical University, Tokyo 160‑8402, Japan
| | - Masato Ogawa
- Department of Biochemistry, Tokyo Medical University, Tokyo 160‑8402, Japan
| | - Masaya Miyazaki
- Department of Biochemistry, Tokyo Medical University, Tokyo 160‑8402, Japan
| | - Kohki Ota
- Department of Biochemistry, Tokyo Medical University, Tokyo 160‑8402, Japan
| | - Hiromi Kazama
- Department of Biochemistry, Tokyo Medical University, Tokyo 160‑8402, Japan
| | - Ayako Hirota
- Department of Biochemistry, Tokyo Medical University, Tokyo 160‑8402, Japan
| | - Naoharu Takano
- Department of Biochemistry, Tokyo Medical University, Tokyo 160‑8402, Japan
| | - Masaki Hiramoto
- Department of Biochemistry, Tokyo Medical University, Tokyo 160‑8402, Japan
| | - Keisuke Miyazawa
- Department of Biochemistry, Tokyo Medical University, Tokyo 160‑8402, Japan
| |
Collapse
|
6
|
Bernardo-Faura M, Rinas M, Wirbel J, Pertsovskaya I, Pliaka V, Messinis DE, Vila G, Sakellaropoulos T, Faigle W, Stridh P, Behrens JR, Olsson T, Martin R, Paul F, Alexopoulos LG, Villoslada P, Saez-Rodriguez J. Prediction of combination therapies based on topological modeling of the immune signaling network in multiple sclerosis. Genome Med 2021; 13:117. [PMID: 34271980 PMCID: PMC8284018 DOI: 10.1186/s13073-021-00925-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2019] [Accepted: 06/14/2021] [Indexed: 11/21/2022] Open
Abstract
Background Multiple sclerosis (MS) is a major health problem, leading to a significant disability and patient suffering. Although chronic activation of the immune system is a hallmark of the disease, its pathogenesis is poorly understood, while current treatments only ameliorate the disease and may produce severe side effects. Methods Here, we applied a network-based modeling approach based on phosphoproteomic data to uncover the differential activation in signaling wiring between healthy donors, untreated patients, and those under different treatments. Based in the patient-specific networks, we aimed to create a new approach to identify drug combinations that revert signaling to a healthy-like state. We performed ex vivo multiplexed phosphoproteomic assays upon perturbations with multiple drugs and ligands in primary immune cells from 169 subjects (MS patients, n=129 and matched healthy controls, n=40). Patients were either untreated or treated with fingolimod, natalizumab, interferon-β, glatiramer acetate, or the experimental therapy epigallocatechin gallate (EGCG). We generated for each donor a dynamic logic model by fitting a bespoke literature-derived network of MS-related pathways to the perturbation data. Last, we developed an approach based on network topology to identify deregulated interactions whose activity could be reverted to a “healthy-like” status by combination therapy. The experimental autoimmune encephalomyelitis (EAE) mouse model of MS was used to validate the prediction of combination therapies. Results Analysis of the models uncovered features of healthy-, disease-, and drug-specific signaling networks. We predicted several combinations with approved MS drugs that could revert signaling to a healthy-like state. Specifically, TGF-β activated kinase 1 (TAK1) kinase, involved in Transforming growth factor β-1 proprotein (TGF-β), Toll-like receptor, B cell receptor, and response to inflammation pathways, was found to be highly deregulated and co-druggable with all MS drugs studied. One of these predicted combinations, fingolimod with a TAK1 inhibitor, was validated in an animal model of MS. Conclusions Our approach based on donor-specific signaling networks enables prediction of targets for combination therapy for MS and other complex diseases. Supplementary Information The online version contains supplementary material available at 10.1186/s13073-021-00925-8.
Collapse
Affiliation(s)
- Marti Bernardo-Faura
- European Molecular Biology Laboratory, European Bioinformatics Institute, Cambridge, UK.,Centre for Research in Agricultural Genomics (CRAG), CSIC-IRTA-UAB-UB, Campus UAB, Bellaterra, Barcelona, Spain
| | - Melanie Rinas
- Joint Research Center for Computational Biomedicine (JRC-COMBINE), Faculty of Medicine, RWTH-Aachen University, Aachen, Germany
| | - Jakob Wirbel
- European Molecular Biology Laboratory, European Bioinformatics Institute, Cambridge, UK.,Joint Research Center for Computational Biomedicine (JRC-COMBINE), Faculty of Medicine, RWTH-Aachen University, Aachen, Germany
| | - Inna Pertsovskaya
- Institut d' Investigacions Biomèdiques August Pi Sunyer (IDIBAPS), Barcelona, Spain
| | - Vicky Pliaka
- School of Mechanical Engineering, National Technical University of Athens, Zografou, Greece
| | | | - Gemma Vila
- Institut d' Investigacions Biomèdiques August Pi Sunyer (IDIBAPS), Barcelona, Spain
| | | | | | - Pernilla Stridh
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Janina R Behrens
- NeuroCure Clinical Research Center and Department of Neurology, Charité University Medicine Berlin, Berlin, Germany
| | - Tomas Olsson
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | | | - Friedemann Paul
- NeuroCure Clinical Research Center and Department of Neurology, Charité University Medicine Berlin, Berlin, Germany
| | - Leonidas G Alexopoulos
- School of Mechanical Engineering, National Technical University of Athens, Zografou, Greece. .,ProtATonce Ltd., Athens, Greece.
| | - Pablo Villoslada
- Institut d' Investigacions Biomèdiques August Pi Sunyer (IDIBAPS), Barcelona, Spain.
| | - Julio Saez-Rodriguez
- European Molecular Biology Laboratory, European Bioinformatics Institute, Cambridge, UK. .,Joint Research Center for Computational Biomedicine (JRC-COMBINE), Faculty of Medicine, RWTH-Aachen University, Aachen, Germany. .,Institute for Computational Biomedicine, Heidelberg University Hospital and Faculty of Medicine, Heidelberg University, Bioquant, Heidelberg, Germany.
| |
Collapse
|
7
|
Yang T, Zha Z, Yang X, Kang Y, Wang X, Tong Y, Zhao X, Wang L, Fan Y. Neuroprotective Effects of Fingolimod Supplement on the Retina and Optic Nerve in the Mouse Model of Experimental Autoimmune Encephalomyelitis. Front Neurosci 2021; 15:663541. [PMID: 33981197 PMCID: PMC8107225 DOI: 10.3389/fnins.2021.663541] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Accepted: 04/06/2021] [Indexed: 01/28/2023] Open
Abstract
Favorable effects exerted by long-term administration of fingolimod therapy in multiple sclerosis (MS) patients have been reported, but sporadic side effects, such as reversible macular edema, also have been recorded. The present study aimed to determine whether fingolimod therapy is beneficial to the visual system in experimental autoimmune encephalomyelitis (EAE) mice. A decrease in demyelination and axon loss in the optic nerve as well as cellular infiltration, especially the recruited macrophages, was observed in EAE with fingolimod treatment. Fingolimod administration diminished hypergliosis of macroglia, including astrocytes and Müller cells in the retina and optic nerve in EAE. Microglia were hyperactivated in the retina and optic nerve in the EAE mice compared to controls, which could be alleviated by fingolimod treatment. Moreover, apoptosis of retinal ganglion cells (RGC) and oligodendrocytes in the optic nerve was significantly reduced with fingolimod treatment compared to that in the untreated EAE mice. These results suggested that fingolimod exerts neuroprotective and anti-inflammatory effects on the retina and optic nerve in a mouse model of EAE. Considering the paradox of favorable and side effects of fingolimod on visual system, we speculate that side effects including macular oedema caused by fingolimod during MS treatment is tendency to be vasogenic rather than hypergliosis in optic nerve and retina which warrants further neuroophthalmological investigation.
Collapse
Affiliation(s)
- Tao Yang
- Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Zheng Zha
- School of Traditional Chinese Medicine, Capital Medical University, Beijing, China
| | - Xiao Yang
- School of Management Science and Engineering, Shandong University of Finance and Economics, Jinan, China
| | - YueZhi Kang
- Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Xin Wang
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Yanping Tong
- Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - XueSong Zhao
- Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Lei Wang
- School of Traditional Chinese Medicine, Capital Medical University, Beijing, China
| | - YongPing Fan
- Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
8
|
Okura I, Kamata M, Asano Y, Mitsui A, Shimizu T, Sato S, Tada Y. Fingolimod ameliorates imiquimod-induced psoriasiform dermatitis by sequestrating interleukin-17-producing ?d T cells in secondary lymph nodes. J Dermatol Sci 2021; 102:116-125. [PMID: 33888401 DOI: 10.1016/j.jdermsci.2021.04.004] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 03/23/2021] [Accepted: 04/13/2021] [Indexed: 11/30/2022]
Abstract
BACKGROUND Psoriasis is a chronic inflammatory skin disease. Interleukin (IL)-17A plays a key role in the pathogenesis of psoriasis. Fingolimod, which is available for the treatment of multiple sclerosis, exerts anti-inflammatory effects by sequestrating inflammatory lymphocytes in secondary lymphoid tissues and the thymus. The effect of fingolimod on psoriasis has not been reported yet. OBJECTIVE Our objectives were to investigate the effect of fingolimod on psoriasis utilizing mice with imiquimod (IMQ)-induced psoriasiform dermatitis, and explore the possibility of fingolimod as a therapeutic agent for psoriasis. METHODS Psoriasiform dermatitis was induced by imiquimod application on murine shaved back skin for six days. Fingolimod prepared in phosphate-buffered saline (PBS), or PBS alone as a control, was administered intraperitoneally daily from days 0 to 5. RESULTS Fingolimod ameliorated IMQ-induced psoriasis dermatitis clinically and histologically. On day 6, the mRNA expression level of IL-17A was lower in the skin of fingolimod-treated mice than in that of PBS-treated mice, whereas it was higher in the inguinal lymph nodes of fingolimod-treated mice than in those of PBS-treated mice. Flow cytometric analyses revealed that fingolimod reduced IL-17A-producing ?d T cells infiltrating into the skin, whereas it increased these cells in the inguinal lymph nodes. Fingolimod inhibited egress of Langerhans cells from the skin to lymph nodes. CONCLUSION Our results demonstrated that fingolimod showed effectiveness for IMQ-induced psoriasiform dermatitis by hindering the emigration of IL-17A-producing ?d T cells from the lymph nodes to the skin, and suggest that fingolimod is a promising candidate for the treatment of psoriasis.
Collapse
Affiliation(s)
- Iori Okura
- Department of Dermatology, Teikyo University School of Medicine, Tokyo, Japan; Pharmaceutical Research Center, Meiji Seika Pharma Co., Ltd., Yokohama, Japan
| | - Masahiro Kamata
- Department of Dermatology, Teikyo University School of Medicine, Tokyo, Japan; Department of Dermatology, University of Tokyo Graduate School of Medicine, Tokyo, Japan.
| | - Yoshihide Asano
- Department of Dermatology, University of Tokyo Graduate School of Medicine, Tokyo, Japan.
| | - Aya Mitsui
- Department of Dermatology, University of Tokyo Graduate School of Medicine, Tokyo, Japan
| | - Teruo Shimizu
- Department of Dermatology, Teikyo University School of Medicine, Tokyo, Japan
| | - Shinichi Sato
- Department of Dermatology, University of Tokyo Graduate School of Medicine, Tokyo, Japan
| | - Yayoi Tada
- Department of Dermatology, Teikyo University School of Medicine, Tokyo, Japan
| |
Collapse
|
9
|
Chiba K. Discovery of fingolimod based on the chemical modification of a natural product from the fungus, Isaria sinclairii. J Antibiot (Tokyo) 2020; 73:666-678. [PMID: 32681100 DOI: 10.1038/s41429-020-0351-0] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Accepted: 06/30/2020] [Indexed: 12/15/2022]
Abstract
Fingolimod is a first-in-class of sphingosine-1-phosphate (S1P) receptor modulator and is widely used a therapeutic drug for multiple sclerosis (MS), autoimmune disease in the central nervous system. About 25 year ago, a natural product, myriocin was isolated from culture broths of the fungus Isaria sinclairii. Myriocin, a rather complex amino acid having three successive asymmetric centers, was found to show a potent immunosuppressive activity in vitro; however, it induced a strong toxicity in vivo. To find out a less toxic immunosuppressive candidate, the chemical structure of myriocin was simplified to a nonchiral symmetric 2-substituted-2-aminoproane-1,3-diol framework. Finally, a highly potent immunosuppressant, fingolimod was found by the extensive chemical modification and pharmacological evaluation using skin allograft model in vivo. Throughout the analyses of the mechanism action of fingolimod, it is revealed that S1P receptor 1 (S1P1) plays an essential role in lymphocyte circulation and that the molecular target of fingolimod is S1P1. Phosphorylated fingolimod acts as a "functional" antagonist at S1P1, modulates lymphocyte circulation, and shows a potent immunosuppressive activity. Fingolimod significantly reduced the relapse rate of MS in the clinical studies and has been approved as a new therapeutic drug for MS in more than 80 countries.
Collapse
Affiliation(s)
- Kenji Chiba
- Research Unit Immunology & Inflammation, Innovative Research Division, Mitsubishi Tanabe Pharma Corporation, 1000, Kamoshida-cho, Aoba-ku, Yokohama, Kanagawa, 227-0033, Japan.
| |
Collapse
|
10
|
Wang Z, Higashikawa K, Yasui H, Kuge Y, Ohno Y, Kihara A, Midori YA, Houkin K, Kawabori M. FTY720 Protects Against Ischemia-Reperfusion Injury by Preventing the Redistribution of Tight Junction Proteins and Decreases Inflammation in the Subacute Phase in an Experimental Stroke Model. Transl Stroke Res 2020; 11:1103-1116. [PMID: 32103462 PMCID: PMC7496052 DOI: 10.1007/s12975-020-00789-x] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Revised: 01/22/2020] [Accepted: 02/12/2020] [Indexed: 12/18/2022]
Abstract
Injury due to brain ischemia followed by reperfusion (I/R) may be an important therapeutic target in the era of thrombectomy. FTY720, a widely known sphingosine-1-phosphate receptor agonist, exerts various neuroprotective effects. The aim of this study was to examine the protective effect of FTY720 with respect to I/R injury, especially focusing on blood-brain barrier (BBB) protection and anti-inflammatory effects. Male rats were subjected to transient ischemia and administered vehicle or 0.5 or 1.5 mg/kg of FTY720 immediately before reperfusion. Positron emission tomography (PET) with [18F]DPA-714 was performed 2 and 9 days after the insult to serially monitor neuroinflammation. Bovine and rat brain microvascular endothelial cells (MVECs) were also subjected to oxygen-glucose deprivation (OGD) and reperfusion, and administered FTY720, phosphorylated-FTY720 (FTY720-P), or their inhibitor. FTY720 dose-dependently reduced cell death, the infarct size, cell death including apoptosis, and inflammation. It also ameliorated BBB disruption and neurological deficits compared to in the vehicle group. PET indicated that FTY720 significantly inhibited the worsening of inflammation in later stages. FTY720-P significantly prevented the intracellular redistribution of tight junction proteins but did not increase their mRNA expression. These results suggest that FTY720 can ameliorate I/R injury by protecting the BBB and regulating neuroinflammation.
Collapse
Affiliation(s)
- Zifeng Wang
- Department of Neurosurgery, Graduate School of Medicine, Hokkaido University, Kita 15, Nishi 7, Kita-ku, Sapporo, Hokkaido, 060-8638, Japan
| | - Kei Higashikawa
- Central Institutes of Isotope Science (Laboratory of Integrated Molecular Imaging, Department of Biomedical Imaging, Graduate School of Biomedical Science and Engineering), Hokkaido University, Sapporo, Hokkaido, Japan
| | - Hironobu Yasui
- Central Institutes of Isotope Science (Laboratory of Integrated Molecular Imaging, Department of Biomedical Imaging, Graduate School of Biomedical Science and Engineering), Hokkaido University, Sapporo, Hokkaido, Japan
| | - Yuji Kuge
- Central Institutes of Isotope Science (Laboratory of Integrated Molecular Imaging, Department of Biomedical Imaging, Graduate School of Biomedical Science and Engineering), Hokkaido University, Sapporo, Hokkaido, Japan
| | - Yusuke Ohno
- Laboratory of Biochemistry, Faculty of Pharmaceutical Sciences, Hokkaido University, Sapporo, Hokkaido, Japan
| | - Akio Kihara
- Laboratory of Biochemistry, Faculty of Pharmaceutical Sciences, Hokkaido University, Sapporo, Hokkaido, Japan
| | - Yenari A Midori
- Department of Neurology, University of California, San Francisco and the San Francisco Veterans Affairs Medical Center, San Francisco, CA, USA
| | - Kiyohiro Houkin
- Department of Neurosurgery, Graduate School of Medicine, Hokkaido University, Kita 15, Nishi 7, Kita-ku, Sapporo, Hokkaido, 060-8638, Japan
| | - Masahito Kawabori
- Department of Neurosurgery, Graduate School of Medicine, Hokkaido University, Kita 15, Nishi 7, Kita-ku, Sapporo, Hokkaido, 060-8638, Japan.
| |
Collapse
|
11
|
Wang Z, Kawabori M, Houkin K. FTY720 (Fingolimod) Ameliorates Brain Injury through Multiple Mechanisms and is a Strong Candidate for Stroke Treatment. Curr Med Chem 2020; 27:2979-2993. [PMID: 31785606 PMCID: PMC7403647 DOI: 10.2174/0929867326666190308133732] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2018] [Revised: 02/12/2019] [Accepted: 02/19/2019] [Indexed: 02/07/2023]
Abstract
FTY720 (Fingolimod) is a known sphingosine-1-phosphate (S1P) receptor agonist that exerts strong anti-inflammatory effects and was approved as the first oral drug for the treatment of multiple sclerosis by the US Food and Drug Administration (FDA) in 2010. FTY720 is mainly associated with unique functional "antagonist" and "agonist" mechanisms. The functional antagonistic mechanism is mediated by the transient down-regulation and degradation of S1P receptors on lymphocytes, which prevents lymphocytes from entering the blood stream from the lymph node. This subsequently results in the development of lymphopenia and reduces lymphocytic inflammation. Functional agonistic mechanisms are executed through S1P receptors expressed on the surface of various cells including neurons, astrocytes, microglia, and blood vessel endothelial cells. These functions might play important roles in regulating anti-apoptotic systems, modulating brain immune and phagocytic activities, preserving the Blood-Brain-Barrier (BBB), and the proliferation of neural precursor cells. Recently, FTY720 have shown receptor-independent effects, including intracellular target bindings and epigenetic modulations. Many researchers have recognized the positive effects of FTY720 and launched basic and clinical experiments to test the use of this agent against stroke. Although the mechanism of FTY720 has not been fully elucidated, its efficacy against cerebral stroke is becoming clear, not only in animal models, but also in ischemic stroke patients through clinical trials. In this article, we review the data obtained from laboratory findings and preliminary clinical trials using FTY720 for stroke treatment.
Collapse
Affiliation(s)
- Zifeng Wang
- Department of Neurosurgery, Hokkaido University Graduate School of Medicine, Sapporo, Hokkaido, Japan
| | - Masahito Kawabori
- Department of Neurosurgery, Hokkaido University Graduate School of Medicine, Sapporo, Hokkaido, Japan
| | - Kiyohiro Houkin
- Department of Neurosurgery, Hokkaido University Graduate School of Medicine, Sapporo, Hokkaido, Japan
| |
Collapse
|
12
|
Amiselimod (MT-1303), a Novel Sphingosine 1-Phosphate Receptor-1 Modulator, Potently Inhibits the Progression of Lupus Nephritis in Two Murine SLE Models. J Immunol Res 2019; 2019:5821589. [PMID: 31930150 PMCID: PMC6942851 DOI: 10.1155/2019/5821589] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Revised: 11/13/2019] [Accepted: 11/29/2019] [Indexed: 11/17/2022] Open
Abstract
Amiselimod (MT-1303) is a novel and selective sphingosine 1-phosphate receptor-1 (S1P1) modulator with a more favorable cardiac safety profile than other S1P1 receptor modulators. In this study, we evaluated the effects of MT-1303 on the progression of lupus nephritis in two well-known murine systemic lupus erythematosus (SLE) models, MRL/lpr and NZBWF1 mice, compared with those of FK506. Daily oral doses of 0.1 and 0.3 mg/kg MT-1303 not only inhibited the development of lupus nephritis when administered before onset in MRL/lpr and NZBWF1 mice but also improved symptoms of lupus nephritis when administered after onset in MRL/lpr mice. Its efficacy in these models was more potent or comparable to that of FK506 (1 and 3 mg/kg). In histological analysis, treatment with MT-1303 inhibited infiltration of T cells into the kidneys, mesangial expansion, and glomerular sclerosis. MT-1303 treatment resulted in a marked reduction in T cells and B cells in the peripheral blood and significantly inhibited increases in the number of plasma cells in the spleen and T cells in the kidneys. In addition, administration of MT-1303 suppressed elevations in serum anti-dsDNA antibody levels in MRL/lpr mice, but not in NZBWF1 mice. Our findings show that MT-1303 exhibits marked therapeutic effects on lupus nephritis in two SLE models, likely by reducing the infiltration of autoreactive T cells into the kidneys. These results suggest that MT-1303 has the potential to be used as a therapeutic agent for patients suffering from SLE, including lupus nephritis.
Collapse
|
13
|
Shimano K, Maeda Y, Kataoka H, Murase M, Mochizuki S, Utsumi H, Oshita K, Sugahara K. Amiselimod (MT-1303), a novel sphingosine 1-phosphate receptor-1 functional antagonist, inhibits progress of chronic colitis induced by transfer of CD4+CD45RBhigh T cells. PLoS One 2019; 14:e0226154. [PMID: 31805144 PMCID: PMC6894856 DOI: 10.1371/journal.pone.0226154] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Accepted: 11/20/2019] [Indexed: 02/07/2023] Open
Abstract
Amiselimod (MT-1303) is a novel sphingosine 1-phosphate receptor-1 (S1P1 receptor) modulator with a more favorable cardiac safety profile than other S1P1 receptor modulators. MT-1303 phosphate (MT-1303-P), an active metabolite of MT-1303, exhibits S1P1 receptor agonism at a lower EC50 value than other S1P1 receptor modulators currently being developed. We aimed to evaluate the efficacy of MT-1303 and its mode of action in chronic colitis using an inflammatory bowel disease (IBD) model. Oral administration of MT-1303 (0.3 mg/kg) once daily for 3 days to mice almost completely abolished S1P1 receptor expression on CD4+ T cells from mesenteric lymph nodes, which corresponded to a marked decrease in CD4+ T cell count in peripheral blood, indicating that MT-1303-P acts as a functional antagonist of the S1P1 receptor. The potential benefit of MT-1303 for IBD was assessed using immunodeficient SCID mice with chronic colitis induced by adoptive transfer of CD4+CD45RBhigh T cells from BALB/c mice. An oral dose of 0.1 and 0.3 mg/kg MT-1303 administered daily one week after the cell transfer inhibited the development of chronic colitis with an efficacy comparable to that of an anti-mTNF-α mAb (250 μg/mouse). In addition, MT-1303 administration significantly reduced the number of infiltrating Th1 and Th17 cells into the lamina propria of the colon in colitis mice. Our results suggest that MT-1303 acts as a functional antagonist of the S1P1 receptor on lymphocytes, regulates lymphocyte trafficking, and inhibits infiltration of colitogenic Th1 and Th17 cells into the colon to inhibit the development of chronic colitis.
Collapse
Affiliation(s)
- Kyoko Shimano
- Sohyaku, Innovative Research Division, Mitsubishi Tanabe Pharma Corporation, Yokohama, Japan
| | - Yasuhiro Maeda
- Sohyaku, Innovative Research Division, Mitsubishi Tanabe Pharma Corporation, Yokohama, Japan
| | - Hirotoshi Kataoka
- Sohyaku, Innovative Research Division, Mitsubishi Tanabe Pharma Corporation, Yokohama, Japan
| | - Mikako Murase
- Sohyaku, Innovative Research Division, Mitsubishi Tanabe Pharma Corporation, Yokohama, Japan
| | - Sachiko Mochizuki
- Sohyaku, Innovative Research Division, Mitsubishi Tanabe Pharma Corporation, Yokohama, Japan
| | - Hiroyuki Utsumi
- Sohyaku, Innovative Research Division, Mitsubishi Tanabe Pharma Corporation, Yokohama, Japan
| | - Koichi Oshita
- Sohyaku, Innovative Research Division, Mitsubishi Tanabe Pharma Corporation, Yokohama, Japan
| | - Kunio Sugahara
- Sohyaku, Innovative Research Division, Mitsubishi Tanabe Pharma Corporation, Yokohama, Japan
- * E-mail:
| |
Collapse
|
14
|
Bauckneht M, Capitanio S, Raffa S, Roccatagliata L, Pardini M, Lapucci C, Marini C, Sambuceti G, Inglese M, Gallo P, Cecchin D, Nobili F, Morbelli S. Molecular imaging of multiple sclerosis: from the clinical demand to novel radiotracers. EJNMMI Radiopharm Chem 2019; 4:6. [PMID: 31659498 PMCID: PMC6453990 DOI: 10.1186/s41181-019-0058-3] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Accepted: 03/21/2019] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Brain PET imaging with different tracers is mainly clinically used in the field of neurodegenerative diseases and brain tumors. In recent years, the potential usefulness of PET has also gained attention in the field of MS. In fact, MS is a complex disease and several processes can be selected as a target for PET imaging. The use of PET with several different tracers has been mainly evaluated in the research setting to investigate disease pathophysiology (i.e. phenotypes, monitoring of progression) or to explore its use a surrogate end-point in clinical trials. RESULTS We have reviewed PET imaging studies in MS in humans and animal models. Tracers have been grouped according to their pathophysiological targets (ie. tracers for myelin kinetic, neuroinflammation, and neurodegeneration). The emerging clinical indication for brain PET imaging in the differential diagnosis of suspected tumefactive demyelinated plaques as well as the clinical potential provided by PET images in view of the recent introduction of PET/MR technology are also addressed. CONCLUSION While several preclinical and fewer clinical studies have shown results, full-scale clinical development programs are needed to translate molecular imaging technologies into a clinical reality that could ideally fit into current precision medicine perspectives.
Collapse
Affiliation(s)
- Matteo Bauckneht
- Nuclear Medicine Unit, IRCCS Ospedale Policlinico San Martino, Largo R. Benzi 10, 16132 Genoa, Italy
| | - Selene Capitanio
- Nuclear Medicine Unit, IRCCS Ospedale Policlinico San Martino, Largo R. Benzi 10, 16132 Genoa, Italy
| | - Stefano Raffa
- Department of Health Sciences (DISSAL), University of Genova, Genoa, Italy
| | - Luca Roccatagliata
- Department of Health Sciences (DISSAL), University of Genova, Genoa, Italy
- Neuroradiology, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Matteo Pardini
- Clinical Neurology, Department of Neuroscience (DINOGMI), University of Genoa, Genoa, Italy
- Clinica Neurologica, IRCCS Ospedale Policlinico, San Martino, Genoa, Italy
| | - Caterina Lapucci
- Clinical Neurology, Department of Neuroscience (DINOGMI), University of Genoa, Genoa, Italy
| | - Cecilia Marini
- Nuclear Medicine Unit, IRCCS Ospedale Policlinico San Martino, Largo R. Benzi 10, 16132 Genoa, Italy
- CNR Institute of Molecular Bioimaging and Physiology, Milan, Italy
| | - Gianmario Sambuceti
- Nuclear Medicine Unit, IRCCS Ospedale Policlinico San Martino, Largo R. Benzi 10, 16132 Genoa, Italy
- Department of Health Sciences (DISSAL), University of Genova, Genoa, Italy
| | - Matilde Inglese
- Clinical Neurology, Department of Neuroscience (DINOGMI), University of Genoa, Genoa, Italy
- Clinica Neurologica, IRCCS Ospedale Policlinico, San Martino, Genoa, Italy
| | - Paolo Gallo
- Multiple Sclerosis Centre of the Veneto Region, Department of Neurosciences DNS, University of Padua, Padua, Italy
| | - Diego Cecchin
- Nuclear Medicine Unit, Department of Medicine-DIMED, Padova University Hospital, Padua, Italy
- Padua Neuroscience Center, University of Padua, Padua, Italy
| | - Flavio Nobili
- Clinical Neurology, Department of Neuroscience (DINOGMI), University of Genoa, Genoa, Italy
- Clinica Neurologica, IRCCS Ospedale Policlinico, San Martino, Genoa, Italy
| | - Silvia Morbelli
- Nuclear Medicine Unit, IRCCS Ospedale Policlinico San Martino, Largo R. Benzi 10, 16132 Genoa, Italy
- Department of Health Sciences (DISSAL), University of Genova, Genoa, Italy
| |
Collapse
|
15
|
Flórido M, Muflihah H, Lin LCW, Xia Y, Sierro F, Palendira M, Feng CG, Bertolino P, Stambas J, Triccas JA, Britton WJ. Pulmonary immunization with a recombinant influenza A virus vaccine induces lung-resident CD4 + memory T cells that are associated with protection against tuberculosis. Mucosal Immunol 2018; 11:1743-1752. [PMID: 30115996 DOI: 10.1038/s41385-018-0065-9] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2018] [Revised: 07/04/2018] [Accepted: 07/05/2018] [Indexed: 02/04/2023]
Abstract
The lung is the primary site of infection with the major human pathogen, Mycobacterium tuberculosis. Effective vaccines against M. tuberculosis must stimulate memory T cells to provide early protection in the lung. Recently, tissue-resident memory T cells (TRM) were found to be phenotypically and transcriptional distinct from circulating memory T cells. Here, we identified M. tuberculosis-specific CD4+ T cells induced by recombinant influenza A viruses (rIAV) vaccines expressing M. tuberculosis peptides that persisted in the lung parenchyma with the phenotypic and transcriptional characteristics of TRMs. To determine if these rIAV-induced CD4+ TRM were protective independent of circulating memory T cells, mice previously immunized with the rIAV vaccine were treated with the sphingosine-1-phosphate receptor modulator, FTY720, prior to and during the first 17 days of M. tuberculosis challenge. This markedly reduced circulating T cells, but had no effect on the frequency of M. tuberculosis-specific CD4+ TRMs in the lung parenchyma or their cytokine response to infection. Importantly, mice immunized with the rIAV vaccine were protected against M. tuberculosis infection even when circulating T cells were profoundly depleted by the treatment. Therefore, pulmonary immunization with the rIAV vaccine stimulates lung-resident CD4+ memory T cells that are associated with early protection against tuberculosis infection.
Collapse
Affiliation(s)
- Manuela Flórido
- Tuberculosis Research Program, Centenary Institute, The University of Sydney, Newtown, NSW, Australia
| | - Heni Muflihah
- Tuberculosis Research Program, Centenary Institute, The University of Sydney, Newtown, NSW, Australia
| | - Leon C W Lin
- Tuberculosis Research Program, Centenary Institute, The University of Sydney, Newtown, NSW, Australia
| | - Yingju Xia
- School of Medicine, Deakin University, Geelong, VIC, Australia
| | - Frederic Sierro
- Liver Immunology Program, Centenary Institute, The University of Sydney, Newtown, NSW, Australia.,Department of Pathology, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
| | - Mainthan Palendira
- Tuberculosis Research Program, Centenary Institute, The University of Sydney, Newtown, NSW, Australia.,Department of Infectious Diseases and Immunology, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
| | - Carl G Feng
- Tuberculosis Research Program, Centenary Institute, The University of Sydney, Newtown, NSW, Australia.,Department of Infectious Diseases and Immunology, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
| | - Patrick Bertolino
- Liver Immunology Program, Centenary Institute, The University of Sydney, Newtown, NSW, Australia.,AW Morrow Gastroenterology and Liver Centre, Royal Prince Alfred Hospital, Camperdown, NSW, Australia
| | - John Stambas
- School of Medicine, Deakin University, Geelong, VIC, Australia
| | - James A Triccas
- Tuberculosis Research Program, Centenary Institute, The University of Sydney, Newtown, NSW, Australia.,Department of Infectious Diseases and Immunology, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
| | - Warwick J Britton
- Tuberculosis Research Program, Centenary Institute, The University of Sydney, Newtown, NSW, Australia. .,Department of Infectious Diseases and Immunology, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia. .,Department of Medicine, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia.
| |
Collapse
|
16
|
Rapamycin and fingolimod modulate Treg/Th17 cells in experimental autoimmune encephalomyelitis by regulating the Akt-mTOR and MAPK/ERK pathways. J Neuroimmunol 2018; 324:26-34. [DOI: 10.1016/j.jneuroim.2018.08.012] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2018] [Revised: 08/24/2018] [Accepted: 08/24/2018] [Indexed: 12/31/2022]
|
17
|
Dillenburg A, Ireland G, Holloway RK, Davies CL, Evans FL, Swire M, Bechler ME, Soong D, Yuen TJ, Su GH, Becher JC, Smith C, Williams A, Miron VE. Activin receptors regulate the oligodendrocyte lineage in health and disease. Acta Neuropathol 2018; 135:887-906. [PMID: 29397421 PMCID: PMC5954071 DOI: 10.1007/s00401-018-1813-3] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2017] [Revised: 01/16/2018] [Accepted: 01/29/2018] [Indexed: 12/12/2022]
Abstract
The most prevalent neurological disorders of myelin include perinatal brain injury leading to cerebral palsy in infants and multiple sclerosis in adults. Although these disorders have distinct etiologies, they share a common neuropathological feature of failed progenitor differentiation into myelin-producing oligodendrocytes and lack of myelin, for which there is an unmet clinical need. Here, we reveal that a molecular pathology common to both disorders is dysregulation of activin receptors and that activin receptor signaling is required for the majority of myelin generation in development and following injury. Using a constitutive conditional knockout of all activin receptor signaling in oligodendrocyte lineage cells, we discovered this signaling to be required for myelination via regulation of oligodendrocyte differentiation and myelin compaction. These processes were found to be dependent on the activin receptor subtype Acvr2a, which is expressed during oligodendrocyte differentiation and axonal ensheathment in development and following myelin injury. During efficient myelin regeneration, Acvr2a upregulation was seen to coincide with downregulation of Acvr2b, a receptor subtype with relatively higher ligand affinity; Acvr2b was shown to be dispensable for activin receptor-driven oligodendrocyte differentiation and its overexpression was sufficient to impair the abovementioned ligand-driven responses. In actively myelinating or remyelinating areas of human perinatal brain injury and multiple sclerosis tissue, respectively, oligodendrocyte lineage cells expressing Acvr2a outnumbered those expressing Acvr2b, whereas in non-repairing lesions Acvr2b+ cells were increased. Thus, we propose that following human white matter injury, this increase in Acvr2b expression would sequester ligand and consequently impair Acvr2a-driven oligodendrocyte differentiation and myelin formation. Our results demonstrate dysregulated activin receptor signaling in common myelin disorders and reveal Acvr2a as a novel therapeutic target for myelin generation following injury across the lifespan.
Collapse
|
18
|
Liu H, Jin H, Yue X, Luo Z, Liu C, Rosenberg AJ, Tu Z. PET Imaging Study of S1PR1 Expression in a Rat Model of Multiple Sclerosis. Mol Imaging Biol 2017; 18:724-32. [PMID: 26975859 DOI: 10.1007/s11307-016-0944-y] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
PURPOSE Upregulation of sphingosine-1-phosphate receptor 1 (S1PR1) expression in multiple sclerosis (MS) lesions is associated with neuroinflammatory response. This study investigated the correlation between neuroinflammation and S1PR1 expression in the spinal cord of an experimental autoimmune encephalomyelitis (EAE) rat model of MS, using the S1PR1 positron emission tomography (PET) radiotracer [(11)C]TZ3321. PROCEDURES MicroPET imaging studies of [(11)C]TZ3321 were performed to measure uptake of [(11)C]TZ3321 in the spinal cord of EAE rats. Immunohistochemical staining was performed to confirm the overexpression of S1PR1 and other inflammatory biomarkers. RESULTS MicroPET imaging demonstrated a 20-30 % increase in [(11)C]TZ3321 uptake in the lumbar spinal cord of EAE rats versus sham controls at 35-60 min post injection. The increased uptake of [(11)C]TZ3321 was correlated with the overexpression of S1PR1 in the lumbar spinal cord of EAE rats that was confirmed by immunohistochemical staining. Upregulated S1PR1 expression was associated with glial cell activation and immune cell infiltration. CONCLUSIONS MicroPET imaging modality with a specific radioligand [(11)C]TZ3321 is able to assess the expression of S1PR1 in EAE rat lumbar spinal cord. This may provide a new approach to the assessment of neuroinflammatory response in MS and other inflammatory diseases.
Collapse
Affiliation(s)
- Hui Liu
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Hongjun Jin
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Xuyi Yue
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Zonghua Luo
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Chunling Liu
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Adam J Rosenberg
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Zhude Tu
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, 63110, USA.
| |
Collapse
|
19
|
Sharma L, Prakash H. Sphingolipids Are Dual Specific Drug Targets for the Management of Pulmonary Infections: Perspective. Front Immunol 2017; 8:378. [PMID: 28400772 PMCID: PMC5372786 DOI: 10.3389/fimmu.2017.00378] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2017] [Accepted: 03/16/2017] [Indexed: 11/13/2022] Open
Abstract
Sphingolipids are the major constituent of the mucus secreted by the cells of epithelial linings of lungs where they maintain the barrier functions and prevent microbial invasion. Sphingolipids are interconvertible, and their primary and secondary metabolites have both structural and functional roles. Out of several sphingolipid metabolites, sphingosine-1 phosphate (S1P) and ceramide are central molecules and decisive for sphingolipid signaling. These are produced by enzymatic activity of sphingosine kinase-1 (SK-1) upon the challenge with either biological or physiological stresses. S1P and ceramide rheostat are important for the progression of various pathologies, which are manifested by inflammatory cascade. S1P is a well-established secondary messenger and associated with various neuronal, metabolic, and inflammatory diseases other than respiratory infections such as Chlamydia pneumoniae, Streptococcus pneumoniae, and Mycobacterium tuberculosis. These pathogens are known to exploit sphingolipid metabolism for their opportunistic survival. Decreased sphingosine kinase activity/S1P content in the lung and peripheral blood of tuberculosis patients clearly indicated a dysregulation of sphingolipid metabolism during infection and suggest that sphingolipid metabolism is important for management of infection by the host. Our previous study has demonstrated that gain of SK-1 activity is important for the maturation of phagolysosomal compartment, innate activation of macrophages, and subsequent control of mycobacterial replication/growth in macrophages. Furthermore, S1P-mediated amelioration of lung pathology and disease severity in TB patients is believed to be mediated by the selective activation or rearrangement of various S1P receptors (S1PR) particularly S1PR2, which has been effective in controlling respiratory fungal pathogens. Therefore, such specificity of S1P-S1PR would be paramount for triggering inflammatory events, subsequent activation, and fostering bactericidal potential in macrophages for the control of TB. In this review, we have discussed and emphasized that sphingolipids may represent effective novel, yet dual specific drug targets for controlling pulmonary infections.
Collapse
Affiliation(s)
- Lalita Sharma
- Laboratory of Translational Medicine, School of Life Sciences, University of Hyderabad , Hyderabad, Telengana , India
| | - Hridayesh Prakash
- Laboratory of Translational Medicine, School of Life Sciences, University of Hyderabad , Hyderabad, Telengana , India
| |
Collapse
|
20
|
Gaire BP, Lee CH, Sapkota A, Lee SY, Chun J, Cho HJ, Nam TG, Choi JW. Identification of Sphingosine 1-Phosphate Receptor Subtype 1 (S1P 1) as a Pathogenic Factor in Transient Focal Cerebral Ischemia. Mol Neurobiol 2017; 55:2320-2332. [PMID: 28343295 DOI: 10.1007/s12035-017-0468-8] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2016] [Accepted: 02/23/2017] [Indexed: 01/06/2023]
Abstract
Medically relevant roles of receptor-mediated sphingosine 1-phosphate (S1P) signaling have become a successful or promising target for multiple sclerosis or cerebral ischemia. Animal-based proof-of-concept validation for the latter is particularly through the neuroprotective efficacy of FTY720, a non-selective S1P receptor modulator, presumably via activation of S1P1. In spite of a clear link between S1P signaling and cerebral ischemia, it remains unknown whether the role of S1P1 is pathogenic or neuroprotective. Here, we investigated the involvement of S1P1 along with its role in cerebral ischemia using a transient middle cerebral artery occlusion ("tMCAO") model. Brain damage following tMCAO, as assessed by brain infarction, neurological deficit score, and neural cell death, was reduced by oral administration of AUY954, a selective S1P1 modulator as a functional antagonist, in a therapeutic paradigm, indicating that S1P1 is a pathogenic mediator rather than a neuroprotective mediator. This pathogenic role of S1P1 in cerebral ischemia was reaffirmed because tMCAO-induced brain damage was reduced by genetic knockdown with an intracerebroventricular microinjection of S1P1 shRNA lentivirus into the brain. Genetic knockdown of S1P1 or AUY954 exposure reduced microglial activation, as assessed by reduction in the number of activated microglia and reversed morphology from amoeboid to ramified, and microglial proliferation in ischemic brain. Its role in microglial activation was recapitulated in lipopolysaccharide-stimulated primary mouse microglia, in which the mRNA expression level of TNF-α and IL-1β, well-known markers for microglial activation, was reduced in microglia transfected with S1P1 siRNA. These data suggest that the pathogenic role of S1P1 is associated with microglial activation in ischemic brain. Additionally, the pathogenic role of S1P1 in cerebral ischemia appears to be associated with the blood-brain barrier disruption and brain-derived neurotrophic factor (BDNF) downregulation. Overall, findings from the current study clearly identify S1P1 signaling as a pathogenic factor in transient focal cerebral ischemia, further implicating S1P1 antagonists including functional antagonists as plausible therapeutic agents for human stroke.
Collapse
Affiliation(s)
| | - Chi-Ho Lee
- Gachon University, Incheon, 406-799, Republic of Korea
| | - Arjun Sapkota
- Gachon University, Incheon, 406-799, Republic of Korea
| | - Sang Yeul Lee
- Department of Pharmacy and Institute of Pharmaceutical Science and Technology, Hanyang University, Ansan, 55 Hanynagdaehak-ro, Sangnok-gu, Ansan, Gyeonggi-do, 15588, Republic of Korea
| | - Jerold Chun
- Department of Molecular Biology, Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA, 92037, USA
| | - Hee Jun Cho
- TherlifEx, Namdong-gu, Incheon, 21653, Republic of Korea
| | - Tae-Gyu Nam
- Department of Pharmacy and Institute of Pharmaceutical Science and Technology, Hanyang University, Ansan, 55 Hanynagdaehak-ro, Sangnok-gu, Ansan, Gyeonggi-do, 15588, Republic of Korea.
| | - Ji Woong Choi
- Gachon University, Incheon, 406-799, Republic of Korea.
| |
Collapse
|
21
|
Martín-Álvarez R, Paúl-Fernández N, Palomo V, Gil C, Martínez A, Mengod G. A preliminary investigation of phoshodiesterase 7 inhibitor VP3.15 as therapeutic agent for the treatment of experimental autoimmune encephalomyelitis mice. J Chem Neuroanat 2017; 80:27-36. [DOI: 10.1016/j.jchemneu.2016.12.001] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2016] [Revised: 11/10/2016] [Accepted: 12/09/2016] [Indexed: 01/08/2023]
|
22
|
Thomas K, Sehr T, Proschmann U, Rodriguez-Leal FA, Haase R, Ziemssen T. Fingolimod additionally acts as immunomodulator focused on the innate immune system beyond its prominent effects on lymphocyte recirculation. J Neuroinflammation 2017; 14:41. [PMID: 28231856 PMCID: PMC5322645 DOI: 10.1186/s12974-017-0817-6] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2016] [Accepted: 02/16/2017] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND Growing evidence emphasizes the relevance of sphingolipids for metabolism and immunity of antigen-presenting cells (APC). APCs are key players in balancing tolerogenic and encephalitogenic responses in immunology. In contrast to the well-known prominent effects of sphingosine-1-phosphate (S1P) on lymphocyte trafficking, modulatory effects on APCs have not been fully characterized. METHODS Frequencies and activation profiles of dendritic cell (DC) subtypes, monocytes, and T cell subsets in 35 multiple sclerosis (MS) patients were evaluated prior and after undergoing fingolimod treatment for up to 24 months. Impact of fingolimod and S1P on maturation and activation profile, pro-inflammatory cytokine release, and phagocytotic capacity was assessed in vitro and ex vivo. Modulation of DC-dependent programming of naïve CD4+ T cells, as well as CD4+ and CD8+ T cell proliferation, was also investigated in vitro and ex vivo. RESULTS Fingolimod increased peripheral slanDC count-CD1+ DC, and monocyte frequencies remained stable. While CD4+ T cell count decreased, ratio of Treg/Th17 significantly increased in fingolimod-treated patients over time. CD83, CD150, and HLADR were all inhibited, but CD86 was upregulated in DCs after incubation in the presence of fingolimod. Fingolimod but not S1P was associated with reduced release of pro-inflammatory cytokines from DCs and monocytes in vitro and ex vivo. Fingolimod also inhibited phagocytic capacity of slanDCs and monocytes. After fingolimod, slanDCs demonstrated reduced potential to induce interferon-gamma-expressing Th1 or IL-17-expressing Th17 cells and DC-dependent T cell proliferation in vitro and in fingolimod-treated patients. CONCLUSIONS We present the first evidence that S1P-directed therapies can act additionally as immunomodulators that decrease the pro-inflammatory capabilities of APCs, which is a crucial element in DC-dependent T cell activation and programming.
Collapse
Affiliation(s)
- Katja Thomas
- Center of Clinical Neuroscience, Department of Neurology, Carl Gustav Carus University Hospital, University of Technology Dresden, Fetscherstr. 74, 01307, Dresden, Germany
| | - Tony Sehr
- Center of Clinical Neuroscience, Department of Neurology, Carl Gustav Carus University Hospital, University of Technology Dresden, Fetscherstr. 74, 01307, Dresden, Germany
| | - Undine Proschmann
- Center of Clinical Neuroscience, Department of Neurology, Carl Gustav Carus University Hospital, University of Technology Dresden, Fetscherstr. 74, 01307, Dresden, Germany
| | - Francisco Alejandro Rodriguez-Leal
- Center of Clinical Neuroscience, Department of Neurology, Carl Gustav Carus University Hospital, University of Technology Dresden, Fetscherstr. 74, 01307, Dresden, Germany
| | - Rocco Haase
- Center of Clinical Neuroscience, Department of Neurology, Carl Gustav Carus University Hospital, University of Technology Dresden, Fetscherstr. 74, 01307, Dresden, Germany
| | - Tjalf Ziemssen
- Center of Clinical Neuroscience, Department of Neurology, Carl Gustav Carus University Hospital, University of Technology Dresden, Fetscherstr. 74, 01307, Dresden, Germany.
| |
Collapse
|
23
|
Bonfiglio T, Olivero G, Merega E, Di Prisco S, Padolecchia C, Grilli M, Milanese M, Di Cesare Mannelli L, Ghelardini C, Bonanno G, Marchi M, Pittaluga A. Prophylactic versus Therapeutic Fingolimod: Restoration of Presynaptic Defects in Mice Suffering from Experimental Autoimmune Encephalomyelitis. PLoS One 2017; 12:e0170825. [PMID: 28125677 PMCID: PMC5268435 DOI: 10.1371/journal.pone.0170825] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2016] [Accepted: 01/11/2017] [Indexed: 11/18/2022] Open
Abstract
Fingolimod, the first oral, disease-modifying therapy for MS, has been recently proposed to modulate glutamate transmission in the central nervous system (CNS) of mice suffering from Experimental Autoimmune Encephalomyelitis (EAE) and in MS patients. Our study aims at investigating whether oral fingolimod recovers presynaptic defects that occur at different stages of disease in the CNS of EAE mice. In vivo prophylactic (0.3 mg/kg for 14 days, from the 7th day post immunization, d.p.i, the drug dissolved in the drinking water) fingolimod significantly reduced the clinical symptoms and the anxiety-related behaviour in EAE mice. Spinal cord inflammation, demyelination and glial cell activation are markers of EAE progression. These signs were ameliorated following oral fingolimod administration. Glutamate exocytosis was shown to be impaired in cortical and spinal cord terminals isolated from EAE mice at 21 ± 1 d.p.i., while GABA alteration emerged only at the spinal cord level. Prophylactic fingolimod recovered these presynaptic defects, restoring altered glutamate and GABA release efficiency. The beneficial effect occurred in a dose-dependent, region-specific manner, since lower (0.1-0.03 mg/kg) doses restored, although to a different extent, synaptic defects in cortical but not spinal cord terminals. A delayed reduction of glutamate, but not of GABA, exocytosis was observed in hippocampal terminals of EAE mice at 35 d.p.i. Therapeutic (0.3 mg/kg, from 21 d.p.i. for 14 days) fingolimod restored glutamate exocytosis in the cortex and in the hippocampus of EAE mice at 35 ± 1 d.p.i. but not in the spinal cord, where also GABAergic defects remained unmodified. These results improve our knowledge of the molecular events accounting for the beneficial effects elicited by fingolimod in demyelinating disorders.
Collapse
MESH Headings
- Administration, Oral
- Animals
- Cerebral Cortex/drug effects
- Cerebral Cortex/immunology
- Cerebral Cortex/pathology
- Dose-Response Relationship, Drug
- Encephalomyelitis, Autoimmune, Experimental/drug therapy
- Encephalomyelitis, Autoimmune, Experimental/immunology
- Encephalomyelitis, Autoimmune, Experimental/pathology
- Encephalomyelitis, Autoimmune, Experimental/prevention & control
- Exocytosis/drug effects
- Female
- Fingolimod Hydrochloride/pharmacology
- Glutamic Acid/metabolism
- Glutamic Acid/pharmacology
- Hippocampus/drug effects
- Hippocampus/immunology
- Hippocampus/pathology
- Immunosuppressive Agents/pharmacology
- Mice
- Mice, Inbred C57BL
- Neuroglia/drug effects
- Neuroglia/immunology
- Neuroglia/pathology
- Organ Specificity
- Spinal Cord/drug effects
- Spinal Cord/immunology
- Spinal Cord/pathology
- Synapses/drug effects
- Synapses/immunology
- Synapses/pathology
- gamma-Aminobutyric Acid/metabolism
- gamma-Aminobutyric Acid/pharmacology
Collapse
Affiliation(s)
- Tommaso Bonfiglio
- Department of Pharmacy, Pharmacology and Toxicology Section, School of Medical and Pharmaceutical Sciences, University of Genoa, Genoa, Italy
| | - Guendalina Olivero
- Department of Pharmacy, Pharmacology and Toxicology Section, School of Medical and Pharmaceutical Sciences, University of Genoa, Genoa, Italy
| | - Elisa Merega
- Department of Pharmacy, Pharmacology and Toxicology Section, School of Medical and Pharmaceutical Sciences, University of Genoa, Genoa, Italy
| | - Silvia Di Prisco
- Department of Pharmacy, Pharmacology and Toxicology Section, School of Medical and Pharmaceutical Sciences, University of Genoa, Genoa, Italy
| | - Cristina Padolecchia
- Department of Pharmacy, Pharmacology and Toxicology Section, School of Medical and Pharmaceutical Sciences, University of Genoa, Genoa, Italy
| | - Massimo Grilli
- Department of Pharmacy, Pharmacology and Toxicology Section, School of Medical and Pharmaceutical Sciences, University of Genoa, Genoa, Italy
- Center of Excellence for Biomedical Research, University of Genoa, Genoa, Italy
| | - Marco Milanese
- Department of Pharmacy, Pharmacology and Toxicology Section, School of Medical and Pharmaceutical Sciences, University of Genoa, Genoa, Italy
- Center of Excellence for Biomedical Research, University of Genoa, Genoa, Italy
| | - Lorenzo Di Cesare Mannelli
- Department of Neuroscience, Psychology, Drug Research and Child Health, Neurofarba, Pharmacology and Toxicology section, University of Florence, Florence, Italy
| | - Carla Ghelardini
- Department of Neuroscience, Psychology, Drug Research and Child Health, Neurofarba, Pharmacology and Toxicology section, University of Florence, Florence, Italy
| | - Giambattista Bonanno
- Department of Pharmacy, Pharmacology and Toxicology Section, School of Medical and Pharmaceutical Sciences, University of Genoa, Genoa, Italy
- Center of Excellence for Biomedical Research, University of Genoa, Genoa, Italy
| | - Mario Marchi
- Department of Pharmacy, Pharmacology and Toxicology Section, School of Medical and Pharmaceutical Sciences, University of Genoa, Genoa, Italy
- Center of Excellence for Biomedical Research, University of Genoa, Genoa, Italy
| | - Anna Pittaluga
- Department of Pharmacy, Pharmacology and Toxicology Section, School of Medical and Pharmaceutical Sciences, University of Genoa, Genoa, Italy
- Center of Excellence for Biomedical Research, University of Genoa, Genoa, Italy
| |
Collapse
|
24
|
Ludwig MD, Turel AP, Zagon IS, McLaughlin PJ. Long-term treatment with low dose naltrexone maintains stable health in patients with multiple sclerosis. Mult Scler J Exp Transl Clin 2016; 2:2055217316672242. [PMID: 28607740 PMCID: PMC5433405 DOI: 10.1177/2055217316672242] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2016] [Accepted: 09/11/2016] [Indexed: 01/11/2023] Open
Abstract
Introduction A retrospective study was conducted on patients at Penn State Hershey Medical Center diagnosed with relapsing–remitting multiple sclerosis between 2006 and 2015. Methodology Laboratory and clinical data collected over this 10-year period were reviewed. Two cohorts of patients were established based on their relapsing–remitting multiple sclerosis therapy at the time of their first visit to Penn State. One group of patients (n = 23) was initially prescribed low dose naltrexone at the time first seen at Hershey. This group was offered low dose naltrexone because of symptoms of fatigue or refusal to take an available disease-modifying therapy. The second group of patients (n = 31) was treated with the glatiramer acetate (Copaxone) and offered low dose naltrexone as an adjunct therapy to their disease-modifying therapy. Results Patient data from visits after 1–50 months post-diagnosis were evaluated in a retrospective manner. Data obtained from patient charts included clinical laboratory values from standard blood tests, timed 25-foot walking trials, and changes in magnetic resonance imaging reports. Statistical analyses between the groups and for each patient over time indicated no significant differences in clinical laboratory values, timed walking, or changes in magnetic resonance imaging. Conclusion These data suggest that the apparently non-toxic, inexpensive, biotherapeutic is safe and if taken alone did not result in an exacerbation of disease symptoms.
Collapse
Affiliation(s)
- Michael D Ludwig
- Department of Neural and Behavioral Sciences, the Pennsylvania State University College of Medicine, USA
| | - Anthony P Turel
- Department of Neurology, The Milton S Hershey Medical Center, USA
| | - Ian S Zagon
- Department of Neural and Behavioral Sciences, the Pennsylvania State University College of Medicine, USA
| | - Patricia J McLaughlin
- Department of Neural and Behavioral Sciences, the Pennsylvania State University College of Medicine, USA
| |
Collapse
|
25
|
Wheeler NA, Fuss B. Extracellular cues influencing oligodendrocyte differentiation and (re)myelination. Exp Neurol 2016; 283:512-30. [PMID: 27016069 PMCID: PMC5010977 DOI: 10.1016/j.expneurol.2016.03.019] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2015] [Revised: 03/03/2016] [Accepted: 03/18/2016] [Indexed: 02/07/2023]
Abstract
There is an increasing number of neurologic disorders found to be associated with loss and/or dysfunction of the CNS myelin sheath, ranging from the classic demyelinating disease, multiple sclerosis, through CNS injury, to neuropsychiatric diseases. The disabling burden of these diseases has sparked a growing interest in gaining a better understanding of the molecular mechanisms regulating the differentiation of the myelinating cells of the CNS, oligodendrocytes (OLGs), and the process of (re)myelination. In this context, the importance of the extracellular milieu is becoming increasingly recognized. Under pathological conditions, changes in inhibitory as well as permissive/promotional cues are thought to lead to an overall extracellular environment that is obstructive for the regeneration of the myelin sheath. Given the general view that remyelination is, even though limited in human, a natural response to demyelination, targeting pathologically 'dysregulated' extracellular cues and their downstream pathways is regarded as a promising approach toward the enhancement of remyelination by endogenous (or if necessary transplanted) OLG progenitor cells. In this review, we will introduce the extracellular cues that have been implicated in the modulation of (re)myelination. These cues can be soluble, part of the extracellular matrix (ECM) or mediators of cell-cell interactions. Their inhibitory and permissive/promotional roles with regard to remyelination as well as their potential for therapeutic intervention will be discussed.
Collapse
Affiliation(s)
- Natalie A Wheeler
- Department of Anatomy and Neurobiology, Virginia Commonwealth University School of Medicine, Richmond, VA 23298, United States
| | - Babette Fuss
- Department of Anatomy and Neurobiology, Virginia Commonwealth University School of Medicine, Richmond, VA 23298, United States.
| |
Collapse
|
26
|
de Bruin NMWJ, Schmitz K, Schiffmann S, Tafferner N, Schmidt M, Jordan H, Häußler A, Tegeder I, Geisslinger G, Parnham MJ. Multiple rodent models and behavioral measures reveal unexpected responses to FTY720 and DMF in experimental autoimmune encephalomyelitis. Behav Brain Res 2015; 300:160-74. [PMID: 26692368 DOI: 10.1016/j.bbr.2015.12.006] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2015] [Revised: 11/30/2015] [Accepted: 12/09/2015] [Indexed: 10/22/2022]
Abstract
Experimental autoimmune encephalomyelitis (EAE) is a widely-used rodent model for multiple sclerosis (MS), but a single model can hardly capture all features of MS. We investigated whether behavioral parameters in addition to clinical motor function scores could be used to assess treatment efficacy during score-free intervals in the relapsing-remitting EAE model in SJL/J mice. We studied the effects of the clinical reference compounds FTY720 (fingolimod, 0.5mg/kg/day) and dimethyl fumarate (DMF, 20-30 mg/kg/day) on clinical scores in several rodent EAE models in order to generate efficacy profiles. SJL/J mice with relapsing-remitting EAE were studied using behavioral tests, including rotarod, gait analysis, locomotor activity and grip strength. SJL/J mice were also examined according to Crawley's sociability and preference for social novelty test. Prophylactic treatment with FTY720 prevented clinical scores in three of the four EAE rodent models: Dark Agouti (DA) and Lewis rats and C57BL/6J mice. Neither prophylactic nor late-therapeutic treatment with FTY720 reduced clinical scores or reversed deficits in the rotarod test in SJL/J mice, but we observed effects on motor functions and sociability in the absence of clinical scores. Prophylactic treatment with FTY720 improved the gait of SJL/J mice whereas late-therapeutic treatment improved manifestations of reduced social (re)cognition or preference for social novelty. DMF was tested in three EAE models and did not improve clinical scores at the dose used. These data indicate that improvements in behavioral deficits can occur in absence of clinical scores, which indicate subtle drug effects and may have translational value for human MS.
Collapse
Affiliation(s)
- N M W J de Bruin
- Fraunhofer Institute for Molecular Biology and Applied Ecology IME, Project Group Translational Medicine & Pharmacology TMP, Theodor-Stern-Kai 7, 60596 Frankfurt am Main, Germany.
| | - K Schmitz
- Pharmazentrum Frankfurt/ZAFES, Institute of Clinical Pharmacology, Goethe-University Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany
| | - S Schiffmann
- Fraunhofer Institute for Molecular Biology and Applied Ecology IME, Project Group Translational Medicine & Pharmacology TMP, Theodor-Stern-Kai 7, 60596 Frankfurt am Main, Germany
| | - N Tafferner
- Fraunhofer Institute for Molecular Biology and Applied Ecology IME, Project Group Translational Medicine & Pharmacology TMP, Theodor-Stern-Kai 7, 60596 Frankfurt am Main, Germany
| | - M Schmidt
- Fraunhofer Institute for Molecular Biology and Applied Ecology IME, Project Group Translational Medicine & Pharmacology TMP, Theodor-Stern-Kai 7, 60596 Frankfurt am Main, Germany
| | - H Jordan
- Fraunhofer Institute for Molecular Biology and Applied Ecology IME, Project Group Translational Medicine & Pharmacology TMP, Theodor-Stern-Kai 7, 60596 Frankfurt am Main, Germany
| | - A Häußler
- Pharmazentrum Frankfurt/ZAFES, Institute of Clinical Pharmacology, Goethe-University Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany
| | - I Tegeder
- Pharmazentrum Frankfurt/ZAFES, Institute of Clinical Pharmacology, Goethe-University Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany
| | - G Geisslinger
- Fraunhofer Institute for Molecular Biology and Applied Ecology IME, Project Group Translational Medicine & Pharmacology TMP, Theodor-Stern-Kai 7, 60596 Frankfurt am Main, Germany; Pharmazentrum Frankfurt/ZAFES, Institute of Clinical Pharmacology, Goethe-University Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany
| | - M J Parnham
- Fraunhofer Institute for Molecular Biology and Applied Ecology IME, Project Group Translational Medicine & Pharmacology TMP, Theodor-Stern-Kai 7, 60596 Frankfurt am Main, Germany
| |
Collapse
|
27
|
Diversity and plasticity of microglial cells in psychiatric and neurological disorders. Pharmacol Ther 2015; 154:21-35. [PMID: 26129625 DOI: 10.1016/j.pharmthera.2015.06.010] [Citation(s) in RCA: 152] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2015] [Accepted: 06/25/2015] [Indexed: 02/07/2023]
|
28
|
Maeda Y, Seki N, Kataoka H, Takemoto K, Utsumi H, Fukunari A, Sugahara K, Chiba K. IL-17-Producing Vγ4+ γδ T Cells Require Sphingosine 1-Phosphate Receptor 1 for Their Egress from the Lymph Nodes under Homeostatic and Inflammatory Conditions. THE JOURNAL OF IMMUNOLOGY 2015; 195:1408-16. [PMID: 26170380 DOI: 10.4049/jimmunol.1500599] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/11/2015] [Accepted: 06/18/2015] [Indexed: 01/04/2023]
Abstract
Conventional αβ T cells require sphingosine 1-phosphate (S1P) receptor 1 (S1P1) for circulation through the lymph nodes (LN); however, it is unclear whether γδ T cells use similar mechanisms. In this study, we found that treatment with fingolimod (FTY720, 1 mg/kg, orally) markedly reduced not only conventional CD4 T cells but also circulating γδ T cells (Vγ4(+) and Vγ4(-) subsets) in the blood of mice. In contrast, IL-17(+)Vγ4(+), IL-17(+)Vγ4(-), and IL-17(-)Vγ4(-) subsets were significantly accumulated in the LN after 6 h of FTY720 treatment. By skin application of a synthetic TLR7/8 agonist, Vγ4(+) γδ T cells (IL-17(+) and IL-17(-) subsets) were accumulated and expanded in the draining LN (DLN), whereas the IL-17(+) subset predominantly migrated to the inflamed skin. FTY720 induced a marked sequestration of IL-17-producing Vγ4(+) γδ T cells in the DLN and inhibited their infiltration into the inflamed skin. Similarly, FTY720 inhibited infiltration of Vγ4(+) γδ T cells into the CNS by their sequestration into the DLN in experimental autoimmune encephalomyelitis. Vγ4(+) γδ T cells expressed a significant level of S1P1 and showed a migratory response toward S1P. FTY720 treatment induced almost complete downregulation of S1P1 expression and S1P responsiveness in Vγ4(+) γδ T cells. Our findings strongly suggest that IL-17-producing Vγ4(+) γδ T cells require S1P1 for their egress from the LN under homeostatic and inflammatory conditions. Consequently, inhibition of S1P1-dependent egress of pathogenic IL-17-producing Vγ4(+) γδ T cells from the DLN may partly contribute the clinical therapeutic effects of FTY720 in relapsing multiple sclerosis.
Collapse
Affiliation(s)
- Yasuhiro Maeda
- Pharmacology Research Laboratories I, Research Division, Mitsubishi Tanabe Pharma Corporation, Kanagawa 227-0033, Japan; and
| | - Noriyasu Seki
- Pharmacology Research Laboratories I, Research Division, Mitsubishi Tanabe Pharma Corporation, Kanagawa 227-0033, Japan; and
| | - Hirotoshi Kataoka
- Pharmacology Research Laboratories I, Research Division, Mitsubishi Tanabe Pharma Corporation, Kanagawa 227-0033, Japan; and
| | - Kana Takemoto
- Safety Research Laboratory, Research Division, Mitsubishi Tanabe Pharma Corporation, Saitama 335-8505, Japan
| | - Hiroyuki Utsumi
- Safety Research Laboratory, Research Division, Mitsubishi Tanabe Pharma Corporation, Saitama 335-8505, Japan
| | - Atsushi Fukunari
- Safety Research Laboratory, Research Division, Mitsubishi Tanabe Pharma Corporation, Saitama 335-8505, Japan
| | - Kunio Sugahara
- Pharmacology Research Laboratories I, Research Division, Mitsubishi Tanabe Pharma Corporation, Kanagawa 227-0033, Japan; and
| | - Kenji Chiba
- Pharmacology Research Laboratories I, Research Division, Mitsubishi Tanabe Pharma Corporation, Kanagawa 227-0033, Japan; and
| |
Collapse
|
29
|
Fujiwara M, Anstadt EJ, Khanna KM, Clark RB. Cbl-b-deficient mice express alterations in trafficking-related molecules but retain sensitivity to the multiple sclerosis therapeutic agent, FTY720. Clin Immunol 2015; 158:103-13. [PMID: 25829233 PMCID: PMC4420730 DOI: 10.1016/j.clim.2015.03.018] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2015] [Revised: 03/08/2015] [Accepted: 03/09/2015] [Indexed: 10/23/2022]
Abstract
The variable response to therapy in multiple sclerosis (MS) suggests a need for personalized approaches based on individual genetic differences. GWAS have linked CBLB gene polymorphisms with MS and recent evidence demonstrated that these polymorphisms can be associated with abnormalities in T cell function and response to interferon-β therapy. Cbl-b is an E3 ubiquitin ligase that regulates T cell activation and Cbl-b-deficient (Cbl-b(-/-)) mice show T cell abnormalities described in MS patients. We now show that Cbl-b(-/-) T cells demonstrate significant lymph node trafficking abnormalities. We thus asked whether the MS-approved drug, FTY720, postulated to trap T cells in lymphoid tissues, is less effective in the context of Cbl-b dysfunction. We now report that FTY720 significantly inhibits EAE in Cbl-b(-/-) mice. Our results newly document a role for Cbl-b in T cell trafficking but suggest nevertheless that MS patients with Cbl-b abnormalities may still be excellent candidates for FTY720 treatment.
Collapse
Affiliation(s)
- Mai Fujiwara
- Department of Immunology, University of Connecticut Health Center, Farmington, CT 06032, USA
| | - Emily J Anstadt
- Department of Immunology, University of Connecticut Health Center, Farmington, CT 06032, USA
| | - Kamal M Khanna
- Department of Immunology, University of Connecticut Health Center, Farmington, CT 06032, USA
| | - Robert B Clark
- Department of Immunology, University of Connecticut Health Center, Farmington, CT 06032, USA; Department of Medicine, University of Connecticut Health Center, Farmington, CT 06032, USA.
| |
Collapse
|
30
|
Song ZY, Yamasaki R, Kawano Y, Sato S, Masaki K, Yoshimura S, Matsuse D, Murai H, Matsushita T, Kira JI. Peripheral blood T cell dynamics predict relapse in multiple sclerosis patients on fingolimod. PLoS One 2015; 10:e0124923. [PMID: 25919001 PMCID: PMC4412716 DOI: 10.1371/journal.pone.0124923] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2014] [Accepted: 03/10/2015] [Indexed: 12/27/2022] Open
Abstract
Background Fingolimod efficiently reduces multiple sclerosis (MS) relapse by inhibiting lymphocyte egress from lymph nodes through down-modulation of sphingosine 1-phosphate (S1P) receptors. We aimed to clarify the alterations in peripheral blood T cell subsets associated with MS relapse on fingolimod. Methods/Principal Findings Blood samples successively collected from 23 relapsing-remitting MS patients before and during fingolimod therapy (0.5 mg/day) for 12 months and 18 healthy controls (HCs) were analysed for T cell subsets by flow cytometry. In MS patients, the percentages of central memory T (CCR7+CD45RO+) cells (TCM) and naïve T (CCR7+CD45RO-) cells decreased significantly, while those of effector memory T (CCR7-CD45RA-) and suppressor precursor T (CD28-) cells increased in both CD4+T and CD8+T cells from 2 weeks to 12 months during fingolimod therapy. The percentages of regulatory T (CD4+CD25highCD127low) cells in CD4+T cells and CCR7-CD45RA+T cells in CD8+T cells also increased significantly. Eight relapsed patients demonstrated greater percentages of CD4+TCM than 15 non-relapsed patients at 3 and 6 months (p=0.0051 and p=0.0088, respectively). The IL17-, IL9-, and IL4-producing CD4+T cell percentages were significantly higher at pre-treatment in MS patients compared with HCs (p<0.01 for all), while the IL17-producing CD4+T cell percentages tended to show a transient increase at 2 weeks of fingolimod therapy (pcorr=0.0834). Conclusions The CD4+TCM percentages at 2 weeks to 12 months during fingolimod therapy are related to relapse.
Collapse
Affiliation(s)
- Zi-Ye Song
- Department of Neurology, Neurological Institute, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Ryo Yamasaki
- Department of Neurological Therapeutics, Neurological Institute, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Yuji Kawano
- Department of Neurology, Neurological Institute, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Shinya Sato
- Department of Neurology, Neurological Institute, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Katsuhisa Masaki
- Department of Neurology, Neurological Institute, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Satoshi Yoshimura
- Department of Neurology, Neurological Institute, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Dai Matsuse
- Department of Neurology, Neurological Institute, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Hiroyuki Murai
- Department of Neurology, Neurological Institute, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Takuya Matsushita
- Department of Neurology, Neurological Institute, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Jun-ichi Kira
- Department of Neurology, Neurological Institute, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
- * E-mail:
| |
Collapse
|
31
|
Pelisch N, Dan T, Ichimura A, Sekiguchi H, Vaughan DE, van Ypersele de Strihou C, Miyata T. Plasminogen Activator Inhibitor-1 Antagonist TM5484 Attenuates Demyelination and Axonal Degeneration in a Mice Model of Multiple Sclerosis. PLoS One 2015; 10:e0124510. [PMID: 25915660 PMCID: PMC4411110 DOI: 10.1371/journal.pone.0124510] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2014] [Accepted: 03/15/2015] [Indexed: 11/20/2022] Open
Abstract
Multiple sclerosis (MS) is characterized by inflammatory demyelination and deposition of fibrinogen in the central nervous system (CNS). Elevated levels of a critical inhibitor of the mammalian fibrinolitic system, plasminogen activator inhibitor 1 (PAI-1) have been demonstrated in human and animal models of MS. In experimental studies that resemble neuroinflammatory disease, PAI-1 deficient mice display preserved neurological structure and function compared to wild type mice, suggesting a link between the fibrinolytic pathway and MS. We previously identified a series of PAI-1 inhibitors on the basis of the 3-dimensional structure of PAI-1 and on virtual screening. These compounds have been reported to provide a number of in vitro and in vivo benefits but none was tested in CNS disease models because of their limited capacity to penetrate the blood-brain barrier (BBB). The existing candidates were therefore optimized to obtain CNS-penetrant compounds. We performed an in vitro screening using a model of BBB and were able to identify a novel, low molecular PAI-1 inhibitor, TM5484, with the highest penetration ratio among all other candidates. Next, we tested the effects on inflammation and demyelination in an experimental allergic encephalomyelitis mice model. Results were compared to either fingolimod or 6α-methylprednisolone. Oral administration of TM5484 from the onset of signs, ameliorates paralysis, attenuated demyelination, and axonal degeneration in the spinal cord of mice. Furthermore, it modulated the expression of brain-derived neurotrophic factor, which plays a protective role in neurons against various pathological insults, and choline acetyltransferase, a marker of neuronal density. Taken together, these results demonstrate the potential benefits of a novel PAI-1 inhibitor, TM5484, in the treatment of MS.
Collapse
Affiliation(s)
- Nicolas Pelisch
- Department of Molecular Medicine and Therapy, United Centers for Advanced Research and Translational Medicine, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
- * E-mail:
| | - Takashi Dan
- Department of Molecular Medicine and Therapy, United Centers for Advanced Research and Translational Medicine, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
| | - Atsuhiko Ichimura
- Department of Molecular Medicine and Therapy, United Centers for Advanced Research and Translational Medicine, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
| | - Hiroki Sekiguchi
- Department of Molecular Medicine and Therapy, United Centers for Advanced Research and Translational Medicine, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
| | - Douglas E. Vaughan
- Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, United States of America
| | | | - Toshio Miyata
- Department of Molecular Medicine and Therapy, United Centers for Advanced Research and Translational Medicine, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
| |
Collapse
|
32
|
McLaughlin PJ, McHugh DP, Magister MJ, Zagon IS. Endogenous opioid inhibition of proliferation of T and B cell subpopulations in response to immunization for experimental autoimmune encephalomyelitis. BMC Immunol 2015; 16:24. [PMID: 25906771 PMCID: PMC4407783 DOI: 10.1186/s12865-015-0093-0] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2014] [Accepted: 04/10/2015] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Experimental autoimmune encephalomyelitis (EAE), an animal model of multiple sclerosis, is induced by immunization of mice with myelin oligodendrocytic glycoprotein (MOG35-55) injections, and after 9 days, mice develop behavioral signs of chronic progressive EAE. Proliferation of T and B cells located in peripheral lymph tissues such as spleen and inguinal lymph nodes of C57BL/6J mice are stimulated. The opioid growth factor-opioid growth factor receptor (OGF-OGFr) axis has been shown to effectively limit progression of chronic EAE when mice are treated at the time of induction or at time of established disease. In addition to repressed behavioral profiles, spinal cord neuropathology is diminished in mice treated with OGF or low dosages of naltrexone (LDN). However, there is little or no information on peripheral lymphocyte dynamics following immunization of mice with MOG antigen and treatment with OGF or LDN. METHODS Six-week old female mice were immunized with MOG35-55 and were injected intraperitoneally with OGF or a low dosage of naltrexone (LDN) beginning at the time of immunization; saline-injected immunized mice served as controls. Normal mice received saline for all injections. Periodically over a 2 week period, spleens and inguinal lymph nodes were removed, total lymphocytes counted, and subpopulations of CD4+ and CD8+ specific T-cells, as well as B lymphocytes, were determined by flow cytometry. On day 15 of treatment, lumbar spinal cord tissue was removed; CNS lymphocytes isolated, and assayed for Th1, Th2, and Th17 markers by flow cytometry. RESULTS Exogenous OGF or endogenous OGF following LDN suppressed T and B lymphocyte proliferation in the spleen and inguinal lymph nodes of MOG-immunized mice. Suppression of peripheral immune cell CD4+ and CD8+ T cell proliferation at 5 and 12 days correlated with reductions in clinical behavior. EAE mice treated with OGF for 15 days displayed elevated Th1 and Th17 cells; no subpopulations of Th2-specific T cells were noted. CONCLUSIONS OGF or LDN repress proliferation of CD4+ and CD8+T cells and B220+ B lymphocytes in the spleen and lymph nodes of immunized mice within a week of immunization. These data provide novel mechanistic pathways underlying the efficacy of OGF and LDN therapy for MS.
Collapse
Affiliation(s)
- Patricia J McLaughlin
- Department of Neural & Behavioral Sciences, Pennsylvania State University College of Medicine, 500 University Drive, MC H109, Hershey, PA, USA.
| | - Daniel P McHugh
- Department of Neural & Behavioral Sciences, Pennsylvania State University College of Medicine, 500 University Drive, MC H109, Hershey, PA, USA.
| | - Marcus J Magister
- Department of Neural & Behavioral Sciences, Pennsylvania State University College of Medicine, 500 University Drive, MC H109, Hershey, PA, USA.
| | - Ian S Zagon
- Department of Neural & Behavioral Sciences, Pennsylvania State University College of Medicine, 500 University Drive, MC H109, Hershey, PA, USA.
| |
Collapse
|
33
|
Luessi F, Kraus S, Trinschek B, Lerch S, Ploen R, Paterka M, Roberg T, Poisa-Beiro L, Klotz L, Wiendl H, Bopp T, Jonuleit H, Jolivel V, Zipp F, Witsch E. FTY720 (fingolimod) treatment tips the balance towards less immunogenic antigen-presenting cells in patients with multiple sclerosis. Mult Scler 2015; 21:1811-22. [PMID: 25732840 DOI: 10.1177/1352458515574895] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2014] [Accepted: 01/26/2015] [Indexed: 11/16/2022]
Abstract
OBJECTIVE We aimed to clarify whether fingolimod has direct effects on antigen-presenting cells in multiple sclerosis patients. METHODS Frequency and phenotype of directly ex vivo dendritic cells and monocytes were analyzed in 43 individuals, including fingolimod-treated and untreated multiple sclerosis patients as well as healthy subjects. These cells were further stimulated with lipopolysaccharide to determine functional effects of fingolimod treatment. RESULTS Absolute numbers of CD1c+ dendritic cells and monocytes were not significantly reduced in fingolimod-treated patients indicating that fingolimod did not block the migration of antigen-presenting cells to peripheral blood. CD86 was upregulated on CD1c+ dendritic cells and thus their activation was not impaired under fingolimod treatment. Quantitative analyses of gene transcription in cells and protein content in supernatants from ex vivo CD1c+ dendritic cells and monocytes, however, showed lower secretion of TNFα, IL1-β and IL-6 upon lipopolysaccharide-stimulation. These results could be matched with CD4+MOG-specific transgenic T cells exhibiting reduced levels of TNFα and IFN-γ but not IL-4 upon stimulation with murine dendritic cells loaded with MOG, when treated with fingolimod. CONCLUSIONS Our data indicate that fingolimod - apart from trapping lymphocytes in lymph nodes - exerts its disease-modulating activity by rebalancing the immune tolerance networks by modulation of antigen-presenting cells.
Collapse
Affiliation(s)
- Felix Luessi
- Department of Neurology, University Medical Center of the Johannes Gutenberg-University of Mainz, Germany
| | - Stefan Kraus
- Department of Neurology, University Medical Center of the Johannes Gutenberg-University of Mainz, Germany
| | - Bettina Trinschek
- Department of Dermatology, University Medical Center of the Johannes Gutenberg-University of Mainz, Germany
| | - Steffen Lerch
- Department of Neurology, University Medical Center of the Johannes Gutenberg-University of Mainz, Germany
| | - Robert Ploen
- Department of Neurology, University Medical Center of the Johannes Gutenberg-University of Mainz, Germany
| | - Magdalena Paterka
- Department of Neurology, University Medical Center of the Johannes Gutenberg-University of Mainz, Germany
| | - Torsten Roberg
- Department of Neurology, University Medical Center of the Johannes Gutenberg-University of Mainz, Germany
| | - Laura Poisa-Beiro
- Department of Neurology, University Medical Center of the Johannes Gutenberg-University of Mainz, Germany
| | - Luisa Klotz
- Department of Neurology, University of Münster, Germany
| | - Heinz Wiendl
- Department of Neurology, University of Münster, Germany
| | - Tobias Bopp
- Institute for Immunology, University Medical Center of the Johannes Gutenberg-University of Mainz, Germany
| | - Helmut Jonuleit
- Department of Dermatology, University Medical Center of the Johannes Gutenberg-University of Mainz, Germany
| | - Valérie Jolivel
- Department of Neurology, University Medical Center of the Johannes Gutenberg-University of Mainz, Germany
| | - Frauke Zipp
- Department of Neurology, University Medical Center of the Johannes Gutenberg-University of Mainz, Germany
| | - Esther Witsch
- Department of Neurology, University Medical Center of the Johannes Gutenberg-University of Mainz, Germany
| |
Collapse
|
34
|
Yoshida Y, Mikami N, Tsuji T, Takada Y, Nakazawa Y, Dan R, Takatsuji M, Fujita T, Tsujikawa K, Kohno T. Mechanism of induction of immune tolerance in experimental autoimmune encephalomyelitis by combination treatment with fingolimod plus pathogenic autoantigen. ACTA ACUST UNITED AC 2014. [DOI: 10.1111/cen3.12140] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Affiliation(s)
- Yuya Yoshida
- Department of Pathological Biochemistry; Faculty of Pharmaceutical Sciences; Setsunan University; Osaka Japan
| | - Norihisa Mikami
- Laboratory of Molecular and Cellular Physiology; Graduate School of Pharmaceutical Sciences; Osaka University; Osaka Japan
| | - Takumi Tsuji
- Department of Pathological Biochemistry; Faculty of Pharmaceutical Sciences; Setsunan University; Osaka Japan
| | - Yuki Takada
- Department of Pathological Biochemistry; Faculty of Pharmaceutical Sciences; Setsunan University; Osaka Japan
| | - Yuka Nakazawa
- Department of Pathological Biochemistry; Faculty of Pharmaceutical Sciences; Setsunan University; Osaka Japan
| | - Rie Dan
- Department of Pathological Biochemistry; Faculty of Pharmaceutical Sciences; Setsunan University; Osaka Japan
| | - Miku Takatsuji
- Laboratory of Molecular and Cellular Physiology; Graduate School of Pharmaceutical Sciences; Osaka University; Osaka Japan
| | - Tetsuro Fujita
- Research Institute for Production and Development; Kyoto Japan
| | - Kazutake Tsujikawa
- Laboratory of Molecular and Cellular Physiology; Graduate School of Pharmaceutical Sciences; Osaka University; Osaka Japan
| | - Takeyuki Kohno
- Department of Pathological Biochemistry; Faculty of Pharmaceutical Sciences; Setsunan University; Osaka Japan
- Research Institute for Production and Development; Kyoto Japan
| |
Collapse
|
35
|
Qian J, Ye Y, Lv L, Zhu C, Ye S. FTY720 attenuates paraquat-induced lung injury in mice. Int Immunopharmacol 2014; 21:426-31. [PMID: 24893116 DOI: 10.1016/j.intimp.2014.05.025] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2014] [Revised: 05/10/2014] [Accepted: 05/21/2014] [Indexed: 10/25/2022]
Abstract
Paraquat (PQ) poisoning, with the lung as a primary target organ, is a devastating disease which irreversibly progresses to diffuse alveolitis followed by extensive lung fibrosis. In the present study, we aimed to investigate the effect of FTY720, an immune modulator, on PQ-induced lung injury in mice. C57BL/6 mice were randomized into four groups: 1) PQ group (n=12): mice was instilled with PQ (30 mg/kg, ip); 2) PQ+FTY720 group (n=12): animals received FTY720 (0.1mg/kg, ip) solution 2h after PQ exposure and twice a week for 4 consecutive weeks; 3) FTY720 group (n=5): FTY720 (0.1mg/kg, ip) was administrated twice a week for 4 consecutive weeks; and 4) Control group (n=10): same volumes of saline were injected. Mice were sacrificed on either day 3 or day 28 for histopathological, biochemical and immunohistochemical analyses of lung damage indicators. We found that FTY720 treatment attenuated PQ-induced acute lung injury and lung fibrosis as evaluated by histopathological changes and Ashcroft score. On day 3, FTY720 administration reduced PQ-induced increases in lung wet weight/body weight (LW/BW), total protein and cytokine levels including interleukin-1β (IL-1β), interleukin-6 (IL-6) and tumor necrosis factor-α (TNF-α) in bronchoalceolar lavage fluid (BALF). On day 28, the expressions of alpha-smooth muscle actin (α-SMA), transforming growth factor-beta (TGF-β) and vascular endothelial growth factor (VEGF) detected by immunohistochemistry, as well as the mRNA levels of α-SMA, Type-I Collagen and Type-III Collagen examined by Real-time PCR were down-regulated after FTY720 treatment. These results indicate that FTY720 could attenuate PQ-induced lung injury, but further investigation is necessary.
Collapse
Affiliation(s)
- Jie Qian
- Department of Emergency Medicine, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai 200127, China
| | - Yan Ye
- Department of Emergency Medicine, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai 200127, China; Department of Rheumatology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai 200001, China
| | - Lixiong Lv
- Department of Emergency Medicine, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai 200127, China
| | - Changqing Zhu
- Department of Emergency Medicine, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai 200127, China.
| | - Shuang Ye
- Department of Rheumatology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai 200001, China.
| |
Collapse
|
36
|
Sheridan GK, Dev KK. Targeting S1P receptors in experimental autoimmune encephalomyelitis in mice improves early deficits in locomotor activity and increases ultrasonic vocalisations. Sci Rep 2014; 4:5051. [PMID: 24851861 PMCID: PMC4031479 DOI: 10.1038/srep05051] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2014] [Accepted: 05/06/2014] [Indexed: 12/31/2022] Open
Abstract
Fingolimod (FTY720) is an oral therapy for relapsing remitting multiple sclerosis (MS) and targets sphingosine 1-phosphate receptors (S1PRs). FTY720 also rescues animals from experimental autoimmune encephalomyelitis (EAE), an animal model of MS. The protective effects of FTY720 in EAE are primarily scored manually by examining weight loss and limb paralysis that begins around 10-12 days after immunisation. To our knowledge, pre-clinical effects of FTY720 on animal behaviour early in EAE have not been explored. Here, we developed an automated behaviour monitoring system to examine the early effects of FTY720 on subtle pre-symptomatic behaviour of mice induced with EAE. Our automated home-cage monitoring system (AHC-MS) enabled non-contact detection of movement and ultrasonic vocalisations (USVs) of mice induced with EAE, thus allowing detection of subtle changes in mouse behaviour before paralysis occurs. Mice receiving FTY720 emit longer USVs and display higher levels of motor activity than vehicle-treated EAE mice before clinical symptoms become apparent. Importantly, this study promotes the 3Rs ethics (replacement, reduction and refinement) in the EAE animal model and may also improve pre-screening of potentially novel MS therapies. In addition, this is the first report showing the early effects of FTY720 in EAE which underscores its protective effects.
Collapse
MESH Headings
- Animals
- Disease Models, Animal
- Encephalomyelitis, Autoimmune, Experimental/metabolism
- Encephalomyelitis, Autoimmune, Experimental/pathology
- Encephalomyelitis, Autoimmune, Experimental/prevention & control
- Female
- Fingolimod Hydrochloride
- Immunosuppressive Agents/pharmacology
- Lysophospholipids/metabolism
- Mice
- Mice, Inbred C57BL
- Motor Activity/drug effects
- Motor Activity/physiology
- Propylene Glycols/pharmacology
- Receptors, Lysosphingolipid/antagonists & inhibitors
- Receptors, Lysosphingolipid/genetics
- Receptors, Lysosphingolipid/metabolism
- Sphingosine/analogs & derivatives
- Sphingosine/metabolism
- Sphingosine/pharmacology
- Vocalization, Animal/drug effects
- Vocalization, Animal/physiology
Collapse
Affiliation(s)
- Graham K. Sheridan
- Drug Development, School of Medicine, Trinity College Dublin, Dublin, Ireland
- Current address: Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge CB2 3DY, UK
| | - Kumlesh K. Dev
- Drug Development, School of Medicine, Trinity College Dublin, Dublin, Ireland
| |
Collapse
|
37
|
Ntranos A, Hall O, Robinson DP, Grishkan IV, Schott JT, Tosi DM, Klein SL, Calabresi PA, Gocke AR. FTY720 impairs CD8 T-cell function independently of the sphingosine-1-phosphate pathway. J Neuroimmunol 2014; 270:13-21. [PMID: 24680062 DOI: 10.1016/j.jneuroim.2014.03.007] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2013] [Revised: 01/24/2014] [Accepted: 03/03/2014] [Indexed: 12/29/2022]
Abstract
Fingolimod (FTY720) is a multiple sclerosis (MS) therapeutic that upon phosphorylation causes the internalization of sphingosine-1-phosphate receptors (S1PR) and traps CCR7+ T-cells in lymph nodes but relatively spares CCR7-effector T-cells. Nonetheless, FTY720-treated patients are more susceptible to viral infections, indicating a CD8 T-cell defect. Thus, the effects of FTY720 on CD8 T-cells were investigated. To this end, we utilized experimental autoimmune encephalomyelitis (EAE) and a murine influenza model. CD8 T-cell trafficking, IFNγ and Granzyme B (GrB) production were assessed by flow cytometry. CD8 T-cell cytotoxic function was assessed in vitro by an LDH release assay. FTY720 not only ameliorated EAE by sequestering T-cells, but also reduced IFNγ and Granzyme B (GrB) in splenic CD8 T-cells. Murine influenza infection was exacerbated and mortality was increased, as FTY720 inhibited CD8 T-cell GrB production and lung infiltration. Remarkably, only the unphosphorylated compound was able to reduce IFNγ and GrB levels in CD8 T-cells and inhibits their cytotoxic function in vitro. The phosphorylated moiety had no effect in vitro, indicating that CD8 T-cell suppression by FTY720 is independent of S1PR modulation. The addition of arachidonic acid rescued CD8 T-cell function, suggesting that this effect may be mediated via inhibition of cytosolic phospholipase A2. Herein, we demonstrate that FTY720 suppresses CD8 T-cells independently of its trafficking effects and S1PR modulation. This provides a novel explanation not only for the increased rate of viral infections in FTY720-treated patients, but also for its efficacy in MS, as CD8 T-cells have emerged as crucial mediators of MS pathogenesis.
Collapse
Affiliation(s)
- Achilles Ntranos
- Department of Neurology, Johns Hopkins University School of Medicine, 600 N. Wolfe St., Baltimore, MD 21287, USA
| | - Olivia Hall
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, The Johns Hopkins Bloomberg School of Public Health, 615 N. Wolfe St., Baltimore, MD 21205, USA
| | - Dionne P Robinson
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, The Johns Hopkins Bloomberg School of Public Health, 615 N. Wolfe St., Baltimore, MD 21205, USA
| | - Inna V Grishkan
- Department of Neurology, Johns Hopkins University School of Medicine, 600 N. Wolfe St., Baltimore, MD 21287, USA
| | - Jason T Schott
- Department of Neurology, Johns Hopkins University School of Medicine, 600 N. Wolfe St., Baltimore, MD 21287, USA
| | - Dominique M Tosi
- Department of Neurology, Johns Hopkins University School of Medicine, 600 N. Wolfe St., Baltimore, MD 21287, USA
| | - Sabra L Klein
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, The Johns Hopkins Bloomberg School of Public Health, 615 N. Wolfe St., Baltimore, MD 21205, USA
| | - Peter A Calabresi
- Department of Neurology, Johns Hopkins University School of Medicine, 600 N. Wolfe St., Baltimore, MD 21287, USA.
| | - Anne R Gocke
- Department of Neurology, Johns Hopkins University School of Medicine, 600 N. Wolfe St., Baltimore, MD 21287, USA.
| |
Collapse
|
38
|
A new phenylpyrazoleanilide, y-320, inhibits interleukin 17 production and ameliorates collagen-induced arthritis in mice and cynomolgus monkeys. Pharmaceuticals (Basel) 2013; 7:1-17. [PMID: 24366113 PMCID: PMC3915191 DOI: 10.3390/ph7010001] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2013] [Revised: 12/16/2013] [Accepted: 12/18/2013] [Indexed: 01/30/2023] Open
Abstract
Interleukin (IL)-15 and IL-17 are thought to play an important role in the pathogenesis of rheumatoid arthritis (RA) because both pro-inflammatory cytokines are found in synovial fluid of RA patients. In this study, we examined the pharmacological profiles of Y-320, a new phenylpyrazoleanilide immunomodulator. Y-320 inhibited IL-17 production by CD4 T cells stimulated with IL-15 with IC50 values of 20 to 60 nM. Oral administration of Y-320 (0.3 to 3 mg/kg) significantly inhibited the development and progression of arthritis and joint destruction with reduction of IL-17 mRNA expression in arthritic joints of type II collagen-induced arthritis (CIA) in DBA/1J mice. Y-320 in combination with anti-murine tumor necrosis factor-α monoclonal antibody showed a synergistic effect on mouse CIA. Moreover, therapeutic treatment with Y-320 (0.3 and 1 mg/kg orally) ameliorated CIA in cynomolgus monkeys. Our results suggest that Y-320, an orally active inhibitor for IL-17 production, provides a useful therapy for RA.
Collapse
|
39
|
Treatment of a relapse-remitting model of multiple sclerosis with opioid growth factor. Brain Res Bull 2013; 98:122-31. [DOI: 10.1016/j.brainresbull.2013.08.001] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2013] [Revised: 07/31/2013] [Accepted: 08/03/2013] [Indexed: 12/30/2022]
|
40
|
Suppression of Experimental Autoimmune Optic Neuritis by the Novel Agent Fingolimod. J Neuroophthalmol 2013; 33:143-8. [DOI: 10.1097/wno.0b013e31828ea2fc] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
41
|
Gebregiworgis T, Massilamany C, Gangaplara A, Thulasingam S, Kolli V, Werth MT, Dodds ED, Steffen D, Reddy J, Powers R. Potential of urinary metabolites for diagnosing multiple sclerosis. ACS Chem Biol 2013; 8:684-90. [PMID: 23369377 PMCID: PMC3631445 DOI: 10.1021/cb300673e] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
A definitive diagnostic test for multiple sclerosis (MS) does not exist; instead physicians use a combination of medical history, magnetic resonance imaging, and cerebrospinal fluid analysis (CSF). Significant effort has been employed to identify biomarkers from CSF to facilitate MS diagnosis; however, none of the proposed biomarkers have been successful to date. Urine is a proven source of metabolite biomarkers and has the potential to be a rapid, noninvasive, inexpensive, and efficient diagnostic tool for various human diseases. Nevertheless, urinary metabolites have not been extensively explored as a source of biomarkers for MS. We demonstrate that urinary metabolites have significant promise for monitoring disease-progression, and response to treatment in MS patients. NMR analysis of urine permitted the identification of metabolites that differentiate experimental autoimmune encephalomyelitis (EAE)-mice (prototypic disease model for MS) from healthy and MS drug-treated EAE mice.
Collapse
Affiliation(s)
- Teklab Gebregiworgis
- Department of Chemistry, University of Nebraska-Lincoln, Lincoln, NE, 68588-0304
| | - Chandirasegaran Massilamany
- School of Veterinary Medicine and Biomedical Sciences, University of Nebraska-Lincoln, Lincoln, NE 68588-0905
| | - Arunakumar Gangaplara
- School of Veterinary Medicine and Biomedical Sciences, University of Nebraska-Lincoln, Lincoln, NE 68588-0905
| | - Sivasubramani Thulasingam
- School of Veterinary Medicine and Biomedical Sciences, University of Nebraska-Lincoln, Lincoln, NE 68588-0905
| | - Venkata Kolli
- Department of Chemistry, University of Nebraska-Lincoln, Lincoln, NE, 68588-0304
| | - Mark T. Werth
- Department of Chemistry, Nebraska Wesleyan University, Lincoln NE 68504
| | - Eric D. Dodds
- Department of Chemistry, University of Nebraska-Lincoln, Lincoln, NE, 68588-0304
| | - David Steffen
- School of Veterinary Medicine and Biomedical Sciences, University of Nebraska-Lincoln, Lincoln, NE 68588-0905
| | - Jay Reddy
- School of Veterinary Medicine and Biomedical Sciences, University of Nebraska-Lincoln, Lincoln, NE 68588-0905
| | - Robert Powers
- Department of Chemistry, University of Nebraska-Lincoln, Lincoln, NE, 68588-0304
| |
Collapse
|
42
|
Murphy RP, Murphy KJ, Pickering M. The development of myelin repair agents for treatment of multiple sclerosis: progress and challenges. Bioengineered 2012; 4:140-6. [PMID: 23147071 DOI: 10.4161/bioe.22835] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Multiple Sclerosis (MS) is an inflammatory demyelinating disorder which affects the central nervous system. Multiple sclerosis treatment has traditionally focused on preventing inflammatory damage to the myelin sheath. Indeed, all currently available disease modifying agents are immunomodulators. However, the limitations of this approach are becoming increasingly clear, leading to the exploration of other potential therapeutic strategies. In particular, targeting the endogenous remyelination system to promote replacement of the lost myelin sheath has shown much promise. As our understanding of remyelination biology advances, the realization of a remyelinating therapeutic comes closer to fruition. In our review, we aim to summarize the limitations of the current immune focused treatment strategy and discuss the potential of remyelination as a new treatment method. Finally, we aim to highlight the challenges in the identification and development of such therapeutics.
Collapse
Affiliation(s)
- Robert P Murphy
- Neurotherapeutics Research Group, UCD School of Biomolecular and Biomedical Science, UCD Conway Institute, University College Dublin, Dublin, Ireland
| | | | | |
Collapse
|
43
|
Discovery of fingolimod, the sphingosine 1-phosphate receptor modulator and its application for the therapy of multiple sclerosis. Future Med Chem 2012; 4:771-81. [PMID: 22530640 DOI: 10.4155/fmc.12.25] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Fingolimod (FTY720) is a first-in-class, orally active, sphingosine 1-phosphate (S1P)-receptor modulator with a structure closely related to sphingosine. The compound was discovered by chemical modification of a natural product, myriocin. Phosphorylated form of FTY720 acts as a functional antagonist at S1P receptor type 1 (S1P(1)), inhibits lymphocyte egress from secondary lymphoid organs and shows immunomodulating effects. Phase III studies in multiple sclerosis demonstrated that oral FTY720 had superior efficacy compared with intramuscular IFN-β1a (AVONEX(®)) with regard to reducing the rate of relapse and the number of inflammatory lesions in the CNS. FTY720 has been approved as a new therapeutic drug for multiple sclerosis in more than 50 countries, including the USA, Japan and some of those in the EU.
Collapse
|
44
|
Chiba K, Yoshii N. [Pharmacological properties and clinical efficacy of fingolimod hydrochloride (Imusera®/Gilenya®) for the treatment of multiple sclerosis]. Nihon Yakurigaku Zasshi 2012; 139:265-74. [PMID: 22728990 DOI: 10.1254/fpj.139.265] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
|
45
|
Badawi AH, Siahaan TJ. Immune modulating peptides for the treatment and suppression of multiple sclerosis. Clin Immunol 2012; 144:127-38. [PMID: 22722227 DOI: 10.1016/j.clim.2012.05.010] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2012] [Revised: 05/25/2012] [Accepted: 05/28/2012] [Indexed: 12/17/2022]
Abstract
Multiple sclerosis (MS) is a neurodegenerative disease in which the immune system recognizes proteins of the myelin sheath as antigenic, thus initiating an inflammatory reaction in the central nervous system. This leads to demyelination of the axons, breakdown of the blood-brain barrier, and lesion formation. Current therapies for the treatment of MS are generally non-specific and weaken the global immune system, thus making the individual susceptible to opportunistic infections. Antigenic peptides and their derivatives are becoming more prevalent for investigation as therapeutic agents for MS because they possess immune-specific characteristics. In addition, other peptides that target vital components of the inflammatory immune response have also been developed. Therefore, the objectives of this review are to (a) summarize the immunological basis for the development of MS, (b) discuss specific and non-specific peptides tested in EAE and in humans, and (c) briefly address some problems and potential solutions with these novel therapies.
Collapse
Affiliation(s)
- Ahmed H Badawi
- Department of Pharmaceutical Chemistry, University of Kansas, Lawrence, KS 66047, USA
| | | |
Collapse
|
46
|
Watson L, Tullus K, Marks SD, Holt RCL, Pilkington C, Beresford MW. Increased serum concentration of sphingosine-1-phosphate in juvenile-onset systemic lupus erythematosus. J Clin Immunol 2012; 32:1019-25. [PMID: 22648459 DOI: 10.1007/s10875-012-9710-3] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2012] [Accepted: 05/22/2012] [Indexed: 10/28/2022]
Abstract
PURPOSE Sphingosine-1-phosphate (S1P) is an active sphingolipid with chemotactic abilities and has been linked to inflammatory mediators and autoimmune disease. The aim of this study was to assess whether children with juvenile-onset systemic lupus erythematosus (JSLE) express increased systemic and/or urinary concentrations of S1P. METHODS A subgroup of patients participating in the UK JSLE Cohort Study, were invited to participate. Cross sectional serum and urine samples were prospectively collected along with demographic and standard clinical data. Results were compared to a cohort of disease controls (Henoch Schonlein Purpura; HSP) and healthy controls (HC). RESULTS The median age of JSLE patients (n = 15) was 13.6 years (7.2-16.9 years). The serum concentrations of S1P in JSLE patients (7.4 uM, IQR 6.3-12.3 uM) were statistically significantly increased when compared to patients with HSP (n = 10; 5.2 uM, IQR 4.0-7.9 uM; p = 0.016) and HCs (n = 10; 3.8 uM, IQR 2.1-5.8 uM; p = 0.003). There was a trend towards increased serum S1P concentrations between patients with active lupus nephritis (n = 8; 8.7 uM, IQR 6.2-15.3 uM) compared to lupus non-nephritis (n = 7; 6.6 uM, IQR 6.3-10.6 uM; p = 0.355). No relationship was found between disease activity markers and S1P. Urine S1P concentrations were no different between JSLE patients (56.0 nM, IQR 40.3-96.6 nM) and HCs (58.7 nM, IQR 0-241.9 nM; p = 0.889). CONCLUSIONS We have demonstrated, for the first time, an increased serum concentration of S1P in a cohort of JSLE patients. These findings highlight a role of S1P in the pathophysiology of JSLE that warrants further investigation.
Collapse
Affiliation(s)
- L Watson
- Department of Women's and Children's Health, Institute of Translational Medicine, University of Liverpool, Alder Hey Children's NHS Foundation Trust Hospital, Eaton Road, Liverpool, L12 2AP, UK.
| | | | | | | | | | | |
Collapse
|
47
|
Sphingosine 1-phosphate receptor 1 as a useful target for treatment of multiple sclerosis. Pharmaceuticals (Basel) 2012; 5:514-28. [PMID: 24281561 PMCID: PMC3763654 DOI: 10.3390/ph5050514] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2012] [Revised: 04/24/2012] [Accepted: 05/15/2012] [Indexed: 12/20/2022] Open
Abstract
Sphingosine 1-phosphate (S1P), a lysophospholipid mediator, is generated from sphingosine by sphingosine kinases and binds five known cell surface receptors. S1P receptor 1 (S1P1) plays an essential role in lymphocyte egress from secondary lymphoid organs (SLO), as evinced by the inability of lymphocytes to exit from the SLO in mice lacking lymphocytic S1P1. Fingolimod hydrochloride (FTY720) is a first-in-class, orally active, S1P receptor modulator with a structure closely related to sphingosine. FTY720 was first synthesized by chemical modification of a natural product, myriocin. FTY720 is effectively converted to an active metabolite, FTY720 phosphate (FTY720-P) by sphingosine kinases. FTY720-P shows high affinity to 4 of the S1P receptors (S1P1, S1P3, S1P4, and S1P5). In particular, FTY720-P strongly induces internalization and degradation of S1P1, inhibits S1P responsiveness of lymphocytes in the SLO, and acts as a functional antagonist at lymphocytic S1P1. Consequently, FTY720 inhibits S1P1-dependent lymphocyte egress from the SLO to decrease circulation of lymphocytes including autoreactive Th17 cells and is highly effective in experimental autoimmune encephalomyelitis (EAE), an animal model of multiple sclerosis (MS). Because FTY720 shows a superior efficacy in relapsing remitting MS patients compared to intramuscular interferon-β-1a (Avonex®), S1P1 is presumed to be a useful target for the therapy of MS.
Collapse
|
48
|
Constantinescu CS, Farooqi N, O'Brien K, Gran B. Experimental autoimmune encephalomyelitis (EAE) as a model for multiple sclerosis (MS). Br J Pharmacol 2012; 164:1079-106. [PMID: 21371012 DOI: 10.1111/j.1476-5381.2011.01302.x] [Citation(s) in RCA: 1085] [Impact Index Per Article: 83.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Experimental autoimmune encephalomyelitis (EAE) is the most commonly used experimental model for the human inflammatory demyelinating disease, multiple sclerosis (MS). EAE is a complex condition in which the interaction between a variety of immunopathological and neuropathological mechanisms leads to an approximation of the key pathological features of MS: inflammation, demyelination, axonal loss and gliosis. The counter-regulatory mechanisms of resolution of inflammation and remyelination also occur in EAE, which, therefore can also serve as a model for these processes. Moreover, EAE is often used as a model of cell-mediated organ-specific autoimmune conditions in general. EAE has a complex neuropharmacology, and many of the drugs that are in current or imminent use in MS have been developed, tested or validated on the basis of EAE studies. There is great heterogeneity in the susceptibility to the induction, the method of induction and the response to various immunological or neuropharmacological interventions, many of which are reviewed here. This makes EAE a very versatile system to use in translational neuro- and immunopharmacology, but the model needs to be tailored to the scientific question being asked. While creating difficulties and underscoring the inherent weaknesses of this model of MS in straightforward translation from EAE to the human disease, this variability also creates an opportunity to explore multiple facets of the immune and neural mechanisms of immune-mediated neuroinflammation and demyelination as well as intrinsic protective mechanisms. This allows the eventual development and preclinical testing of a wide range of potential therapeutic interventions.
Collapse
Affiliation(s)
- Cris S Constantinescu
- Division of Clinical Neurology, School of Clinical Sciences, University of Nottingham, Queen's Medical Centre, Nottingham, UK.
| | | | | | | |
Collapse
|
49
|
Chiba K, Kataoka H, Seki N, Maeda Y, Sugahara K. Fingolimod (FTY720), the Sphingosine 1-Phosphate Receptor Modulator, as a New Therapeutic Drug in Multiple Sclerosis. Inflamm Regen 2011. [DOI: 10.2492/inflammregen.31.167] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
|