Siponimod (BAF312) prevents synaptic neurodegeneration in experimental multiple sclerosis

Antonietta Gentile, Alessandra Musella, Silvia Bullitta, Diego Fresegna, Francesca De Vito, Roberta Fantozzi, Eleonora Piras, Francesca Gargano, Giovanna Borsellino, Luca Battistini, Anna Schubart, Georgia Mandolesi, Diego Centonze, Antonietta Gentile, Alessandra Musella, Silvia Bullitta, Diego Fresegna, Francesca De Vito, Roberta Fantozzi, Eleonora Piras, Francesca Gargano, Giovanna Borsellino, Luca Battistini, Anna Schubart, Georgia Mandolesi, Diego Centonze

Abstract

Background: Data from multiple sclerosis (MS) and the MS rodent model, experimental autoimmune encephalomyelitis (EAE), highlighted an inflammation-dependent synaptopathy at the basis of the neurodegenerative damage causing irreversible disability in these disorders. This synaptopathy is characterized by an imbalance between glutamatergic and GABAergic transmission and has been proposed to be a potential therapeutic target. Siponimod (BAF312), a selective sphingosine 1-phosphate1,5 receptor modulator, is currently under investigation in a clinical trial in secondary progressive MS patients. We investigated whether siponimod, in addition to its peripheral immune modulation, may exert direct neuroprotective effects in the central nervous system (CNS) of mice with chronic progressive EAE.

Methods: Minipumps allowing continuous intracerebroventricular (icv) infusion of siponimod for 4 weeks were implanted into C57BL/6 mice subjected to MOG35-55-induced EAE. Electrophysiology, immunohistochemistry, western blot, qPCR experiments, and peripheral lymphocyte counts were performed. In addition, the effect of siponimod on activated microglia was assessed in vitro to confirm the direct effect of the drug on CNS-resident immune cells.

Results: Siponimod administration (0.45 μg/day) induced a significant beneficial effect on EAE clinical scores with minimal effect on peripheral lymphocyte counts. Siponimod rescued defective GABAergic transmission in the striatum of EAE, without correcting the EAE-induced alterations of glutamatergic transmission. We observed a significant attenuation of astrogliosis and microgliosis together with reduced lymphocyte infiltration in the striatum of EAE mice treated with siponimod. Interestingly, siponimod reduced the release of IL-6 and RANTES from activated microglial cells in vitro, which might explain the reduced lymphocyte infiltration. Furthermore, the loss of parvalbumin-positive (PV+) GABAergic interneurons typical of EAE brains was rescued by siponimod treatment, providing a plausible explanation of the selective effects of this drug on inhibitory synaptic transmission.

Conclusions: Altogether, our results show that siponimod has neuroprotective effects in the CNS of EAE mice, which are likely independent of its peripheral immune effect, suggesting that this drug could be effective in limiting neurodegenerative pathological processes in MS.

Keywords: Experimental autoimmune encephalomyelitis; GABA; Neurodegeneration; Parvalbumin neuron; Striatum; Synaptic transmission.

Figures

Fig. 1
Fig. 1
Intracerebroventricular administration of 0.45 μg/day siponimod attenuates EAE motor deficits without inducing peripheral lymphocyte depletion. a Schematic representation of the experimental design used in the study. Minipump implantation and siponimod/vehicle release preceded the induction of EAE by 1 week. All examinations were performed on animals during the peak of the symptomatic phase of the disease 18–24 dpi, both in the peripheral blood samples and in the striatum of the animals. b CD3+ lymphocytes were counted in the peripheral blood of EAE mice receiving vehicle or different siponimod dosages: significant reduction was observed for the 4.5 μg/day dosage. Siponimod 4.5 μg/day vs vehicle ##p < 0.01 unpaired T test; siponimod 0.45 μg/day vs vehicle p > 0.05 unpaired T test; siponimod 0.225 μg/day vs vehicle p > 0.05 unpaired T test. c Siponimod concentrations were determined in peripheral blood samples of EAE mice receiving intracerebroventricular release of different siponimod dosages: the highest concentration (4.5 μg/day) used was significantly higher compared to both 0.450 μg/day and 0.225 μg/day, while there were no statistically differences between the other two dosages. Tukey’s post hoc ***p < 0.001. d Representative clinical course of EAE mice treated with three different siponimod dosages: EAE disease progression is strongly affected by 4.5 μg/day dosage during the symptomatic phase of the disease and significantly attenuated by 0.45 μg/day concentration in the dpi range of 18–24. Daily statistical significance was evaluated by non-parametric Mann-Whitney test
Fig. 2
Fig. 2
CNS-directed delivery of siponimod reduces astrogliosis in the EAE striatum. a Confocal microscopy images depict the striatum of EAE-vehicle and siponimod mice stained for the astrocyte marker GFAP (green fluorescence, counterstained with DAPI, gray). Expression of GFAP is markedly reduced in EAE-siponimod striatum compared to EAE-vehicle striatum (scale bar: 100 μm). The images are representative of stained sections from three animals per groups from two immunizations. The panel in b shows western blot comparing EAE-siponimod and EAE-vehicle striatal extracts probed for anti-GFAP antibody. Densitometric analysis of the bands in b’ reveals reduced GFAP content, relative to β-actin, in EAE-siponimod lysates compared to EAE-vehicle. WB data are normalized to EAE-vehicle values. Unpaired T test was used for two-group analysis. Values are means ± SEM, *p < 0.05
Fig. 3
Fig. 3
Attenuated microgliosis and reduced lymphocyte infiltration characterizes the striatum of EAE mice treated with siponimod. a Double immunostaining of coronal striatal sections (in gray DAPI nuclei) showing expression of Iba1-positive microglia/macrophage cells (red) and of CD3 (green) in EAE-siponimod and EAE-vehicle mice. Only few lymphocytes could be detected in the striatum of mice treated with siponimod, and microgliosis is visibly reduced in the same animals. Scale bar: 100 μm. b The levels of IBA1 and CD3 mRNA were quantified by qRT-PCR in the striatum of EAE-siponimod versus EAE-vehicle mice using β-actin as internal control. The histogram shows that both IBA1 and CD3 mRNA were significantly reduced in the striatum of siponimod-treated mice. Statistical significance was evaluated by unpaired T test: *p < 0.05
Fig. 4
Fig. 4
In vitro siponimod application to activated BV2 microglial cell reduces IL-6 and RANTES release. BV2 cells were pre-treated with siponimod prior to stimulation with TNF. The levels of secreted IL-6 (a) and RANTES (b) were measured by Luminex assay on collected media. Application of siponimod to non-activated cells does not modulate the secretion of both molecules. Data were expressed as picograms per milliliter (pg/ml) and were analyzed by one-way ANOVA, followed by Tukey’s post hoc test. *p < 0.05, **p < 0.01, ***p < 0.001
Fig. 5
Fig. 5
Siponimod treatment corrects GABAergic defects in EAE striatum. a The glutamatergic transmission was recorded from MSNs of EAE-vehicle and EAE-siponimod mice. The analysis of the increased kinetic properties (rise time, decay time, and half width) of sEPSCs showed that there were no differences between the two groups (unpaired T test, p > 0.05). a’ The frequencies of the glutamatergic currents, increased in EAE striatum, were not corrected by siponimod (unpaired T test, *p > 0.05). a” Electrophysiological traces representative of glutamatergic currents recorded from MSNs of EAE-vehicle and EAE-siponimod mice. b Siponimod restores normal GABAergic frequencies in the striatum of acute phase EAE mice (unpaired T test,*p < 0.05). b’ Electrophysiological traces representative of GABAergic currents recorded from MSNs of EAE-vehicle and EAE-siponimod mice. c The frequencies of the GABAergic currents, reduced in EAE striatum, were completely rescued after 1 h of siponimod incubation in EAE slices (unpaired T test, **p < 0.01)
Fig. 6
Fig. 6
Brain infusion of siponimod promotes the survival of PV+ GABAergic interneurons during the course of EAE. a Low magnification images (×5 objective) of coronal striatal sections from control CFA-vehicle, EAE-vehicle, and EAE-siponimod showing area selected for PV counting on serial sections (dorsal striatum) and DAB-immunolabeled interneurons (scale bar: 200 μm): at high magnification, the density of PV+ cells is visibly reduced in EAE-vehicle compared to that in CFA-vehicle with a partial recovery in EAE-siponimod slices. Yellow arrows indicate PV-labeled cells (scale bar: 20 μm). b Histogram represents the quantitative stereological counts of PV+ cells performed on striatal slices: the intracerebroventricular treatment with siponimod significantly increases the total number of PV+ cells in the dorsal left striatum of EAE mice. Data are expressed as mean ± S.E.M. Statistical comparisons showed in the graph were calculated with Newman-Keuls post hoc: CFA-vehicle vs EAE-vehicle **p < 0.01; CFA-vehicle vs EAE-siponimod p > 0.05; EAE-vehicle vs EAE-siponimod*p < 0.05

References

    1. Lublin FD, Reingold SC, Cohen JA, Cutter GR, Sørensen PS, Thompson AJ, et al. Defining the clinical course of multiple sclerosis: the 2013 revisions. Neurology. 2014;83:278–286. doi: 10.1212/WNL.0000000000000560.
    1. Calabrese M, Magliozzi R, Ciccarelli O, Geurts JJ, Reynolds R, Martin R. Exploring the origins of grey matter damage in multiple sclerosis. Nat Rev Neurosci. 2015;16:147–158. doi: 10.1038/nrn3900.
    1. Salvetti M, Landsman D, Schwarz-Lam P, Comi G, Thompson AJ, Fox RJ. Progressive MS: from pathophysiology to drug discovery. Mult Scler. 2015;21:1376–1384. doi: 10.1177/1352458515603802.
    1. Fyfe I. In the news: ocrelizumab excites ECTRIMS. Nat Rev Neurol. 2015;11:667.
    1. Ciccarelli O, Barkhof F, Bodini B, De Stefano N, Golay X, Nicolay K, et al. Pathogenesis of multiple sclerosis: insights from molecular and metabolic imaging. Lancet Neurol. 2014;13:807–822. doi: 10.1016/S1474-4422(14)70101-2.
    1. Rossi S, Muzio L, De Chiara V, Grasselli G, Musella A, Musumeci G, et al. Impaired striatal GABA transmission in experimental autoimmune encephalomyelitis. Brain Behav Immun. 2011;25:947–956. doi: 10.1016/j.bbi.2010.10.004.
    1. Mandolesi G, Grasselli G, Musella A, Gentile A, Musumeci G, Sepman H, et al. GABAergic signaling and connectivity on Purkinje cells are impaired in experimental autoimmune encephalomyelitis. Neurobiol Dis. 2012;46:414–424. doi: 10.1016/j.nbd.2012.02.005.
    1. Mandolesi G, Musella A, Gentile A, Grasselli G, Haji N, Sepman H, et al. Interleukin-1β alters glutamate transmission at purkinje cell synapses in a mouse model of multiple sclerosis. J Neurosci. 2013;33:12105–12121. doi: 10.1523/JNEUROSCI.5369-12.2013.
    1. Nisticò R, Mango D, Mandolesi G, Piccinin S, Berretta N, Pignatelli M, et al. Inflammation subverts hippocampal synaptic plasticity in experimental multiple sclerosis. PLoS One. 2013;8:e54666. doi: 10.1371/journal.pone.0054666.
    1. Mori F, Nisticò R, Mandolesi G, Piccinin S, Mango D, Kusayanagi H, et al. Interleukin-1β promotes long-term potentiation in patients with multiple sclerosis. Neuromolecular Med. 2014;16:38–51. doi: 10.1007/s12017-013-8249-7.
    1. Mandolesi G, Gentile A, Musella A, Fresegna D, De Vito F, Bullitta S, et al. Synaptopathy connects inflammation and neurodegeneration in multiple sclerosis. Nat Rev Neurol. 2015;11:711–724. doi: 10.1038/nrneurol.2015.222.
    1. Plaut GS. Effectiveness of amantadine in reducing relapses in multiple sclerosis. J R Soc Med. 1987;80:91–93.
    1. Cutter NC, Scott DD, Johnson JC, Whiteneck G. Gabapentin effect on spasticity in multiple sclerosis: a placebo-controlled, randomized trial. Arch Phys Med Rehabil. 2000;81:164–169. doi: 10.1016/S0003-9993(00)90135-7.
    1. Pitt D, Werner P, Raine CS. Glutamate excitotoxicity in a model of multiple sclerosis. Nat Med. 2000;6:67–70. doi: 10.1038/71555.
    1. Centonze D, Muzio L, Rossi S, Cavasinni F, De Chiara V, Bergami A, et al. Inflammation triggers synaptic alteration and degeneration in experimental autoimmune encephalomyelitis. J Neurosci. 2009;29:3442–3452. doi: 10.1523/JNEUROSCI.5804-08.2009.
    1. Bhat R, Axtell R, Mitra A, Miranda M, Lock C, Tsien RW, et al. Inhibitory role for GABA in autoimmune inflammation. Proc Natl Acad Sci U S A. 2010;107:2580–2585. doi: 10.1073/pnas.0915139107.
    1. Subei AM, Cohen JA. Sphingosine 1-phosphate receptor modulators in multiple sclerosis. CNS Drugs. 2015;29:565–575. doi: 10.1007/s40263-015-0261-z.
    1. Selmaj K, Li DK, Hartung HP, Hemmer B, Kappos L, Freedman MS, et al. Siponimod for patients with relapsing-remitting multiple sclerosis (BOLD): an adaptive, dose-ranging, randomised, phase 2 study. Lancet Neurol. 2013;12:756–767. doi: 10.1016/S1474-4422(13)70102-9.
    1. Gergely P, Nuesslein-Hildesheim B, Guerini D, Brinkmann V, Traebert M, Bruns C, et al. The selective sphingosine 1-phosphate receptor modulator BAF312 redirects lymphocyte distribution and hasspecies-specific effects on heart rate. Br J Pharmacol. 2012;167:1035–1047. doi: 10.1111/j.1476-5381.2012.02061.x.
    1. Shakeri-Nejad K, Aslanis V, Veldandi UK, Mooney L, Pezous N, Brendani B, et al. Effects of therapeutic and supratherapeutic doses of siponimod (BAF312) on cardiac repolarization in healthy subjects. Clin Ther. 2015;37:2489–2505. doi: 10.1016/j.clinthera.2015.09.006.
    1. Groves A, Kihara Y, Chun J. Fingolimod: direct CNS effects of sphingosine 1-phosphate (S1P) receptor modulation and implications in multiple sclerosis therapy. J Neurol Sci. 2013;328:9–18. doi: 10.1016/j.jns.2013.02.011.
    1. Gentile A, Fresegna D, Federici M, Musella A, Rizzo FR, Sepman H, et al. Dopaminergic dysfunction is associated with IL-1β-dependent mood alterations in experimental autoimmune encephalomyelitis. Neurobiol Dis. 2015;74:347–358. doi: 10.1016/j.nbd.2014.11.022.
    1. Compston A, Coles A. Multiple sclerosis. Lancet. 2008;372:1502–1517. doi: 10.1016/S0140-6736(08)61620-7.
    1. Muzio L, Martino G, Furlan R. Multifaceted aspects of inflammation in multiple sclerosis: the role of microglia. J Neuroimmunol. 2007;191:39–44. doi: 10.1016/j.jneuroim.2007.09.016.
    1. Ajami B, Bennett JL, Krieger C, McNagny KM, Rossi FM. Infiltrating monocytes trigger EAE progression, but do not contribute to the resident microglia pool. Nat Neurosci. 2011;14:1142–1149. doi: 10.1038/nn.2887.
    1. Moreno M, Bannerman P, Ma J, Guo F, Miers L, Soulika AM, et al. Conditional ablation of astroglial CCL2 suppresses CNS accumulation of M1 macrophages and preserves axons in mice with MOG peptide EAE. J Neurosci. 2014;34:8175–8185. doi: 10.1523/JNEUROSCI.1137-14.2014.
    1. Grasselli G, Rossi S, Musella A, Gentile A, Loizzo S, Muzio L, et al. Abnormal NMDA receptor function exacerbates experimental autoimmune encephalomyelitis. Br J Pharmacol. 2013;168:502–517. doi: 10.1111/j.1476-5381.2012.02178.x.
    1. Eugster HP, Frei K, Kopf M, Lassmann H, Fontana A. IL-6-deficient mice resist myelin oligodendrocyte glycoprotein-induced autoimmune encephalomyelitis. Eur J Immunol. 1998;28:2178–2187. doi: 10.1002/(SICI)1521-4141(199807)28:07<2178::AID-IMMU2178>;2-D.
    1. Sørensen TL, Tani M, Jensen J, Pierce V, Lucchinetti C, Folcik VA, et al. Expression of specific chemokines and chemokine receptors in the central nervous system of multiple sclerosis patients. J Clin Invest. 1999;103:807–815. doi: 10.1172/JCI5150.
    1. Baranzini SE, Elfstrom C, Chang SY, Butunoi C, Murray R, Higuchi R, et al. Transcriptional analysis of multiple sclerosis brain lesions reveals a complex pattern of cytokine expression. J Immunol. 2000;165:6576–6582. doi: 10.4049/jimmunol.165.11.6576.
    1. dos Santos AC, Barsante MM, Arantes RM, Bernard CC, Teixeira MM, et al. CCL2 and CCL5 mediate leukocyte adhesion in experimental autoimmune encephalomyelitis—an intravital microscopic study. J Neuroimmunol. 2005;162:122–129. doi: 10.1016/j.jneuroim.2005.01.020.
    1. Furlan R, Rovaris M, Martinelli Boneschi F, Khademi M, Bergami A, et al. Immunological patterns identifying disease course and evolution in multiple sclerosis patients. J Neuroimmunol. 2005;165:192–200. doi: 10.1016/j.jneuroim.2005.04.012.
    1. Kawaguchi Y, Wilson CJ, Augood SJ, Emson PC. Striatal interneurones: chemical, physiological and morphological characterization. Trends Neurosci. 1995;18:527–535. doi: 10.1016/0166-2236(95)98374-8.
    1. Gustafson N, Gireesh-Dharmaraj E, Czubayko U, Blackwell KT, Plenz D. A comparative voltage and current-clamp analysis of feedback and feedforward synaptic transmission in the striatal microcircuit in vitro. J Neurophysiol. 2006;95:737–752. doi: 10.1152/jn.00802.2005.
    1. De Stefano N, Matthews PM, Filippi M, Agosta F, De Luca M, et al. Evidence of early cortical atrophy in MS: relevance to white matter changes and disability. Neurology. 2003;60:1157–1162. doi: 10.1212/01.WNL.0000055926.69643.03.
    1. Roosendaal SD, Bendfeldt K, Vrenken H, Polman CH, Borgwardt S, et al. Grey matter volume in a large cohort of MS patients: relation to MRI parameters and disability. Mult Scler. 2011;17:1098–1106. doi: 10.1177/1352458511404916.
    1. Jackson SJ, Giovannoni G, Baker D. Fingolimod modulates microglial activation to augment markers of remyelination. J Neuroinflammation. 2011;8:76. doi: 10.1186/1742-2094-8-76.
    1. Colombo E, Di Dario M, Capitolo E, Chaabane L, Newcombe J, Martino G, et al. Fingolimod may support neuroprotection via blockade of astrocyte nitric oxide. Ann Neurol. 2014;76:325–337. doi: 10.1002/ana.24217.
    1. Haji N, Mandolesi G, Gentile A, Sacchetti L, Fresegna D, Rossi S, et al. TNF-α-mediated anxiety in a mouse model of multiple sclerosis. Exp Neurol. 2012;237:296–303. doi: 10.1016/j.expneurol.2012.07.010.
    1. Gentile A, De Vito F, Fresegna D, Musella A, Buttari F, Bullitta S, et al. Exploring the role of microglia in mood disorders associated with experimental multiple sclerosis. Front Cell Neurosci. 2015;9:243. doi: 10.3389/fncel.2015.00243.
    1. Uzawa A, Mori M, Uchida T, Masuda H, Ohtani R, Kuwabara S. Increased levels of CSF CD59 in neuromyelitis optica and multiple sclerosis. Clin Chim Acta. 2016;453:131–3. doi: 10.1016/j.cca.2015.12.013.
    1. Maimone D, Guazzi GC, Annunziata P. IL-6 detection in multiple sclerosis brain. J Neurol Sci. 1997;146(1):59–65. doi: 10.1016/S0022-510X(96)00283-3.
    1. Szczuciński A, Losy J. CCL5, CXCL10 and CXCL11 chemokines in patients with active and stable relapsing-remitting multiple sclerosis. Neuroimmunomodulation. 2011;18(1):67–72. doi: 10.1159/000317394.
    1. Mori F, Nisticò R, Nicoletti CG, Zagaglia S, Mandolesi G, Piccinin S et al. RANTES correlates with inflammatory activity and synaptic excitability in multiple sclerosis. Mult Scler. 2016. doi:10.1177/ 1352458515621796.
    1. Rossi S, Lo Giudice T, De Chiara V, Musella A, Studer V, Motta C, et al. Oral fingolimod rescues the functional deficits of synapses in experimental autoimmune encephalomyelitis. Br J Pharmacol. 2012;165:861–869. doi: 10.1111/j.1476-5381.2011.01579.x.
    1. Gentile A, Rossi S, Studer V, Motta C, De Chiara V, Musella A, et al. Glatiramer acetate protects against inflammatory synaptopathy in experimental autoimmune encephalomyelitis. J Neuroimmune Pharmacol. 2013;8:651–663. doi: 10.1007/s11481-013-9436-x.
    1. Musella A, Mandolesi G, Gentile A, Rossi S, Studer V, Motta C, et al. Cladribine interferes with IL-1β synaptic effects in experimental multiple sclerosis. J Neuroimmunol. 2013;264:8–13. doi: 10.1016/j.jneuroim.2013.08.009.
    1. Clements RJ, McDonough J, Freeman EJ. Distribution of parvalbumin and calretinin immunoreactive interneurons in motorcortex from multiple sclerosis post-mortem tissue. Exp Brain Res. 2008;187:459–465. doi: 10.1007/s00221-008-1317-9.
    1. Ji MH, Qiu LL, Tang H, Ju LS, Sun XR, Zhang H, et al. Sepsis-induced selective parvalbumin interneuron phenotype loss and cognitive impairments may be mediated by NADPH oxidase 2 activation in mice. J Neuroinflammation. 2015;12:182. doi: 10.1186/s12974-015-0401-x.
    1. Czeh B, Simon M, van der Hart MG, Schmelting B, Hesselink MB, Fuchs E. Chronic stress decreases the number of parvalbumin-immunoreactive interneurons in the hippocampus: prevention by treatment with a substance P receptor (NK1) antagonist. Neuropsychopharmacology. 2005;30:67–79. doi: 10.1038/sj.npp.1300581.
    1. Allen SJ, Watson JJ, Shoemark DK, Barua NU, Patel NK. GDNF, NGF and BDNF as therapeutic options for neurodegeneration. Pharmacol Ther. 2013;138:155–175. doi: 10.1016/j.pharmthera.2013.01.004.
    1. Cawley N, Solanky BS, Muhlert N, Tur C, Edden RA, Wheeler-Kingshott CA, et al. Reduced gamma-aminobutyric acid concentration is associated with physical disability in progressive multiple sclerosis. Brain. 2015;138:2584–2595. doi: 10.1093/brain/awv209.
    1. Haber SN. The place of dopamine in the cortico-basal ganglia circuit. Neuroscience. 2014;282C:248–257. doi: 10.1016/j.neuroscience.2014.10.008.
    1. Tao G, Datta S, He R, Nelson F, Wolinsky JS, Narayana PA. Deep gray matter atrophy in multiple sclerosis: a tensor based morphometry. J Neurol Sci. 2009;282:39–46. doi: 10.1016/j.jns.2008.12.035.

Source: PubMed

3
S'abonner