Sphingosine 1-phosphate receptor modulation suppresses pathogenic astrocyte activation and chronic progressive CNS inflammation

Significance Secondary progressive multiple sclerosis (SPMS) inflicts severe and irreversible disability on the affected individuals. Astrocytes are thought to play a central role in the pathogenesis of SPMS. Here, we demonstrate that Sphingosine-1-receptor (S1PR) modulation suppresses pathogenic astrocyte activation and disease progression in an animal model of SPMS. Using functional in vitro assays, we defined direct effects of S1PR modulation on murine and human astrocytes, as well as astrocyte-mediated effects on microglia and proinflammatory monocytes. Finally, in unbiased transcriptome-wide studies on human astrocytes, we identified candidate targets for the modulation of astrocyte function in SPMS. Collectively, this study sheds light on the pathogenesis of SPMS and evaluates the therapeutic value of S1PR modulation in an animal model of SPMS. Multiple sclerosis (MS) is an autoimmune inflammatory demyelinating disease of the CNS that causes disability in young adults as a result of the irreversible accumulation of neurological deficits. Although there are potent disease-modifying agents for its initial relapsing-remitting phase, these therapies show limited efficacy in secondary progressive MS (SPMS). Thus, there is an unmet clinical need for the identification of disease mechanisms and potential therapeutic approaches for SPMS. Here, we show that the sphingosine 1-phosphate receptor (S1PR) modulator fingolimod (FTY720) ameliorated chronic progressive experimental autoimmune encephalomyelitis in nonobese diabetic mice, an experimental model that resembles several aspects of SPMS, including neurodegeneration and disease progression driven by the innate immune response in the CNS. Indeed, S1PR modulation by FTY720 in murine and human astrocytes suppressed neurodegeneration-promoting mechanisms mediated by astrocytes, microglia, and CNS-infiltrating proinflammatory monocytes. Genome-wide studies showed that FTY720 suppresses transcriptional programs associated with the promotion of disease progression by astrocytes. The study of the molecular mechanisms controlling these transcriptional modules may open new avenues for the development of therapeutic strategies for progressive MS.

[1]  J. Antel,et al.  Sphingosine-1-Phosphate Receptors in the Central Nervous and Immune Systems. , 2016, Current drug targets.

[2]  X. Montalban,et al.  Safety and Efficacy of Siponimod (BAF312) in Patients With Relapsing-Remitting Multiple Sclerosis: Dose-Blinded, Randomized Extension of the Phase 2 BOLD Study. , 2016, JAMA neurology.

[3]  J. East,et al.  Sphingosine 1-phosphate signaling impacts lymphocyte migration, inflammation and infection. , 2016, Pathogens and disease.

[4]  M. Han,et al.  Sphingosine-1-Phosphate (S1P) and S1P Signaling Pathway: Therapeutic Targets in Autoimmunity and Inflammation , 2016, Drugs.

[5]  H. Stark,et al.  Sphingosine kinase 2 deficient mice exhibit reduced experimental autoimmune encephalomyelitis: Resistance to FTY720 but not ST-968 treatments , 2016, Neuropharmacology.

[6]  M. Haghani,et al.  Fingolimod (FTY720) improves hippocampal synaptic plasticity and memory deficit in rats following focal cerebral ischemia , 2016, Brain Research Bulletin.

[7]  Ludwig Kappos,et al.  Oral fingolimod in primary progressive multiple sclerosis (INFORMS): a phase 3, randomised, double-blind, placebo-controlled trial , 2016, The Lancet.

[8]  B. Engelhardt,et al.  Immune cell trafficking across the barriers of the central nervous system in multiple sclerosis and stroke. , 2016, Biochimica et biophysica acta.

[9]  J. Correale,et al.  Sphingosine 1-phosphate signaling in astrocytes: Implications for progressive multiple sclerosis , 2016, Journal of the Neurological Sciences.

[10]  K. Dev,et al.  The dual S1PR1/S1PR5 drug BAF312 (Siponimod) attenuates demyelination in organotypic slice cultures , 2016, Journal of Neuroinflammation.

[11]  K. Takabe,et al.  Sphingosine-1-Phosphate Signaling in Immune Cells and Inflammation: Roles and Therapeutic Potential , 2016, Mediators of inflammation.

[12]  C. Cooper,et al.  The emerging role of FTY720 (Fingolimod) in cancer treatment , 2016, Oncotarget.

[13]  P. Xiong,et al.  Interleukin-33 is released in spinal cord and suppresses experimental autoimmune encephalomyelitis in mice , 2015, Neuroscience.

[14]  A. Anderson,et al.  The Non-Obese Diabetic Mouse Strain as a Model to Study CD8+ T Cell Function in Relapsing and Progressive Multiple Sclerosis , 2015, Front. Immunol..

[15]  C. Trebst,et al.  Effect of FTY720-phosphate on the expression of inflammation-associated molecules in astrocytes in vitro. , 2015, Molecular medicine reports.

[16]  J. Goldman,et al.  Astrocyte pathology in Alexander disease causes a marked inflammatory environment , 2015, Acta Neuropathologica.

[17]  Control of autoimmune CNS inflammation by astrocytes , 2015, Seminars in Immunopathology.

[18]  E. Briard,et al.  MS565: A SPECT Tracer for Evaluating the Brain Penetration of BAF312 (Siponimod) , 2015, ChemMedChem.

[19]  M. Sofroniew Astrocyte barriers to neurotoxic inflammation , 2015, Nature Reviews Neuroscience.

[20]  B. Trapp,et al.  Astrocyte response to IFN-γ limits IL-6-mediated microglia activation and progressive autoimmune encephalomyelitis , 2015, Journal of Neuroinflammation.

[21]  R. Proia,et al.  Emerging biology of sphingosine-1-phosphate: its role in pathogenesis and therapy. , 2015, The Journal of clinical investigation.

[22]  N. J. Allen,et al.  Astrocytes Control Synapse Formation, Function, and Elimination. , 2015, Cold Spring Harbor perspectives in biology.

[23]  B. Trapp,et al.  Pathological mechanisms in progressive multiple sclerosis , 2015, The Lancet Neurology.

[24]  J. Orian,et al.  Modelling MS: Chronic-Relapsing EAE in the NOD/Lt Mouse Strain. , 2015, Current topics in behavioral neurosciences.

[25]  H. Lassmann Mechanisms of white matter damage in multiple sclerosis , 2014, Glia.

[26]  H. Weiner,et al.  Regulation of astrocyte activation by glycolipids drives chronic CNS inflammation , 2014, Nature Medicine.

[27]  Y. Ao,et al.  Astrocyte CCL2 sustains immune cell infiltration in chronic experimental autoimmune encephalomyelitis , 2014, Journal of Neuroimmunology.

[28]  M. Nedergaard,et al.  White matter astrocytes in health and disease , 2014, Neuroscience.

[29]  J. Newcombe,et al.  Fingolimod may support neuroprotection via blockade of astrocyte nitric oxide , 2014, Annals of neurology.

[30]  W. Pfeilschifter,et al.  Fingolimod for the treatment of neurological diseases—state of play and future perspectives , 2014, Front. Cell. Neurosci..

[31]  T. Hla,et al.  Sphingosine‐1‐phosphate receptor 1 signalling in T cells: trafficking and beyond , 2014, Immunology.

[32]  D. Pleasure,et al.  Conditional Ablation of Astroglial CCL2 Suppresses CNS Accumulation of M1 Macrophages and Preserves Axons in Mice with MOG Peptide EAE , 2014, The Journal of Neuroscience.

[33]  Sarah Spiegel,et al.  Sphingolipid metabolites in inflammatory disease , 2014, Nature.

[34]  Ludwig Kappos,et al.  Safety and efficacy of fingolimod in patients with relapsing-remitting multiple sclerosis (FREEDOMS II): a double-blind, randomised, placebo-controlled, phase 3 trial , 2014, The Lancet Neurology.

[35]  F. Nicoletti,et al.  Molecular pharmacodynamics of new oral drugs used in the treatment of multiple sclerosis , 2014, Drug design, development and therapy.

[36]  N. Sibson,et al.  Investigation of immune and CNS-mediated effects of fingolimod in the focal delayed-type hypersensitivity multiple sclerosis model , 2014, Neuropharmacology.

[37]  S. Gygi,et al.  Identification of a Unique TGF-β Dependent Molecular and Functional Signature in Microglia , 2013, Nature Neuroscience.

[38]  Marja-Leena Linne,et al.  Astrocyte-neuron interactions: from experimental research-based models to translational medicine. , 2014, Progress in molecular biology and translational science.

[39]  K. J. Murphy,et al.  Neuron–glia crosstalk in health and disease: fractalkine and CX3CR1 take centre stage , 2013, Open Biology.

[40]  T. Luedde,et al.  A new type of microglia gene targeting shows TAK1 to be pivotal in CNS autoimmune inflammation , 2013, Nature Neuroscience.

[41]  J. Goverman,et al.  Modeling the heterogeneity of multiple sclerosis in animals. , 2013, Trends in immunology.

[42]  Yasuyuki Kihara,et al.  Fingolimod: Direct CNS effects of sphingosine 1-phosphate (S1P) receptor modulation and implications in multiple sclerosis therapy , 2013, Journal of the Neurological Sciences.

[43]  C. Brosnan,et al.  The astrocyte in multiple sclerosis revisited , 2013, Glia.

[44]  A. Ahmadiani,et al.  FTY720 (Fingolimod) Attenuates Beta-amyloid Peptide (Aβ42)-Induced Impairment of Spatial Learning and Memory in Rats , 2013, Journal of Molecular Neuroscience.

[45]  Jeffrey A. Cohen,et al.  Fingolimod Therapy for Multiple Sclerosis , 2013, Seminars in Neurology.

[46]  N. Gray,et al.  Discovery of BAF312 (Siponimod), a Potent and Selective S1P Receptor Modulator. , 2013, ACS medicinal chemistry letters.

[47]  P. Séguéla,et al.  Dual effects of daily FTY720 on human astrocytes in vitro: relevance for neuroinflammation , 2013, Journal of Neuroinflammation.

[48]  J. Lünemann,et al.  The initiation and prevention of multiple sclerosis , 2012, Nature Reviews Neurology.

[49]  Hans Lassmann,et al.  Progressive multiple sclerosis: pathology and pathogenesis , 2012, Nature Reviews Neurology.

[50]  M. Milovanovic,et al.  IL‐33 attenuates EAE by suppressing IL‐17 and IFN‐γ production and inducing alternatively activated macrophages , 2012, European journal of immunology.

[51]  Kevin W Eliceiri,et al.  NIH Image to ImageJ: 25 years of image analysis , 2012, Nature Methods.

[52]  K. Dev,et al.  S1P1 receptor subtype inhibits demyelination and regulates chemokine release in cerebellar slice cultures , 2012, Glia.

[53]  F. Rossi,et al.  Infiltrating monocytes trigger EAE progression, but do not contribute to the resident microglia pool , 2011, Nature Neuroscience.

[54]  S. Ludwin,et al.  Neurobiological effects of sphingosine 1‐phosphate receptor modulation in the cuprizone model , 2011, FASEB journal : official publication of the Federation of American Societies for Experimental Biology.

[55]  Volker Brinkmann,et al.  Sphingosine 1-phosphate (S1P) , 2011, Neurology.

[56]  L. Kappos,et al.  Clinical immunology of the sphingosine 1-phosphate receptor modulator fingolimod (FTY720) in multiple sclerosis , 2011, Neurology.

[57]  F. Barkhof,et al.  Future clinical challenges in multiple sclerosis , 2011, Neurology.

[58]  D. Herr,et al.  FTY720 (fingolimod) efficacy in an animal model of multiple sclerosis requires astrocyte sphingosine 1-phosphate receptor 1 (S1P1) modulation , 2010, Proceedings of the National Academy of Sciences.

[59]  J. Chun,et al.  Neurological S1P signaling as an emerging mechanism of action of oral FTY720 (Fingolimod) in multiple sclerosis , 2010, Archives of pharmacal research.

[60]  S. Ludwin,et al.  Fingolimod (FTY720) enhances remyelination following demyelination of organotypic cerebellar slices. , 2010, The American journal of pathology.

[61]  Ludwig Kappos,et al.  A placebo-controlled trial of oral fingolimod in relapsing multiple sclerosis. , 2010, The New England journal of medicine.

[62]  H. Hartung,et al.  Mechanism of Action of Oral Fingolimod (FTY720) in Multiple Sclerosis , 2010, Clinical neuropharmacology.

[63]  V. Brinkmann FTY720 (fingolimod) in Multiple Sclerosis: therapeutic effects in the immune and the central nervous system , 2009, British journal of pharmacology.

[64]  A. Mildner,et al.  CCR2+Ly-6Chi monocytes are crucial for the effector phase of autoimmunity in the central nervous system. , 2009, Brain : a journal of neurology.

[65]  H. Weiner,et al.  Toll-like receptor 2 and poly(ADP-ribose) polymerase 1 promote central nervous system neuroinflammation in progressive EAE , 2009, Nature Immunology.

[66]  C. A. Foster,et al.  FTY720 Rescue Therapy in the Dark Agouti Rat Model of Experimental Autoimmune Encephalomyelitis: Expression of Central Nervous System Genes and Reversal of Blood‐Brain‐Barrier Damage , 2009, Brain pathology.

[67]  A. Mildner,et al.  CCR 2 + Ly-6 Chi monocytes are crucial for the effector phase of autoimmunity in the central nervous system , 2009 .

[68]  H. Weiner,et al.  Reversal of axonal loss and disability in a mouse model of progressive multiple sclerosis. , 2008, The Journal of clinical investigation.

[69]  B. Rollins,et al.  Absence of Monocyte Chemoattractant Protein 1 in Mice Leads to Decreased Local Macrophage Recruitment and Antigen-Specific T Helper Cell Type 1 Immune Response in Experimental Autoimmune Encephalomyelitis , 2001, The Journal of experimental medicine.

[70]  H. Weiner,et al.  Resistance to Experimental Autoimmune Encephalomyelitis in Mice Lacking the Cc Chemokine Receptor (Ccr2) , 2000, The Journal of experimental medicine.