Is It worth Considering Circulating microRNAs in Multiple Sclerosis?

Ferdinand Jagot, Nathalie Davoust, Ferdinand Jagot, Nathalie Davoust

Abstract

New evidence has highlighted that miRNA production and trafficking can be dysregulated in both autoimmmune and neurological disorders. Multiple sclerosis (MS) in particular is an autoimmune pathology leading to neurodegeneration. Profiling studies performed on cells derived from MS patients have described a dysregulated network of miRNAs in both immune and neural cells. Interestingly, new evidence has emerged showing that circulating miRNAs are also dysregulated in MS body fluids, including plasma/serum and cerebrospinal fluid. This review summarizes the current scientific theories on the function of this altered circulating miRNA network. It builds up new insights about miRNA transfer mechanisms including extracellular vesicle trafficking involved in cell-to-cell communication and the possible physiopathological functions of these transfers in MS. Finally, this review proposes that monitoring altered miRNA expression levels could serve as a potential biomarker read-out of MS subtype and severity.

Keywords: biomarkers; circulating miRNAs; demyelination; exosomes; extracellular vesicles; inflammation; multiple sclerosis; relapses.

Figures

Figure 1
Figure 1
Overlapping between dysregulated miRNAs in plasma and lymphocytes or in plasma and the CNS of MS patients. Dysregulated miRNAs from plasma and lymphocytes (left panel) or from plasma and the CNS (right panel) were either identical (overlapping area) or not (single area). Data were compiled from miRNA profiling studies performed on plasma (, –11, 18), immune cells [B (–22) or T cells (, –26)], and the CNS (astrocytes, oligodendrocytes, brain endothelial cells, whole brain lesions, and whole brain) (, –30). Briefly, we selected dysregulated miRNAs from microarray profiling studies, filtered out miRNAs with non-significant variation of the expression level, and highlighted commonly dysregulated miRNAs. All overlapping miRNAs are listed and those written in white color are dysregulated in at least three compartments, including plasma and the CNS; miR, miRNA.
Figure 2
Figure 2
Major pathways for extracellular vesicle biogenesis and for miRNA incorporation into exosomes. (1) The exosomal pathway includes MVB formation, exosome budding into MVB lumen, and exocytosis-mediated exosomal release. (a) Ubiquitinated RISC- and ESCRT-dependent incorporation of miRNAs into exosomes: model from Gibbings and colleagues (35). (b) Main pathway of miRNA biogenesis detailed in the Section “Introduction.” (2) The microvesicular pathway: miRNA uptake by microvesicles remains to be clarified. (3) The apoptotic pathway: apoptotic cells release apoptotic bodies containing fragment of the nucleus and putative miRNAs.
Figure 3
Figure 3
Functions of dysregulated cellular and circulating miRNAs in MS/EAE physiopathology. Myelin-derived peptides reach the lymph node as free autoimmune-prone antigens or associated with antigen-presenting cells. It results in B cell and T cell activation and migration of these cells across a porous brain blood barrier. Unbalanced differentiation toward proinflammatory T cell subsets Th1 and Th17 is amplified by cellular miR-155 and miR-326 upregulation and by cell non-autonomous transfer of these miRNAs between T cells through putative EVs. In the CNS, miR-155 participates to microglia-mediated inflammation/neurodegeneration. Brain inflammation is also aggravated by miR-155-mediated decrease of CD47, driving vulnerability of neural cells (evidence being for astrocytes) toward microglia-mediated phagocytosis. miRNAs are depicted as red dashes. EVs appear in the color of the producing cell and contain a single effective miRNA for simplification. HEV, high endothelial venules.
Figure 4
Figure 4
Model of the cell non-autonomous effect of circulating miRNAs. Exacerbated T cell differentiation (evidence being mainly for Th17 cells) might be a feature of the transition from remission to relapses (3). The initial state (remission) would depict poorly differentiated T cells, whereas it ends up during relapses with exaggerated T cell differentiation and activation. The transition from remission to relapses also correlates with increased dysregulation of miRNAs (3, 5) that would drive inflammatory T cell differentiation and activation. The model of cell non-autonomous effects of miRNAs consists in the transfer through EVs of T cell differentiation-driving miRNAs. The cause of initial miRNA dysregulation remains unknown. Light blue, dark blue, and yellow cells depict Th1, Th17, and non-differentiated Th0 cells, respectively.

References

    1. Filipowicz W, Bhattacharyya SN, Sonenberg N. Mechanisms of post-transcriptional regulation by microRNAs: are the answers in sight? Nat Rev Genet (2008) 9:102–14.10.1038/nrg2290
    1. Winter J, Jung S, Keller S, Gregory RI, Diederichs S. Many roads to maturity: microRNA biogenesis pathways and their regulation. Nat Cell Biol (2009) 11:228–34.10.1038/ncb0309-228
    1. Du C, Liu C, Kang J, Zhao G, Ye Z, Huang S, et al. microRNA miR-326 regulates TH-17 differentiation and is associated with the pathogenesis of multiple sclerosis. Nat Immunol (2009) 10:1252–9.10.1038/ni.1798
    1. Junker A, Krumbholz M, Eisele S, Mohan H, Augstein F, Bittner R, et al. microRNA profiling of multiple sclerosis lesions identifies modulators of the regulatory protein CD47. Brain (2009) 132:3342–52.10.1093/brain/awp300
    1. Zhang J, Cheng Y, Cui W, Li M, Li B, Guo L. microRNA-155 modulates Th1 and Th17 cell differentiation and is associated with multiple sclerosis and experimental autoimmune encephalomyelitis. J Neuroimmunol (2014) 266:56–63.10.1016/j.jneuroim.2013.09.019
    1. Murugaiyan G, da Cunha AP, Ajay AK, Joller N, Garo LP, Kumaradevan S, et al. microRNA-21 promotes Th17 differentiation and mediates experimental autoimmune encephalomyelitis. J Clin Invest (2015) 125:1069–80.10.1172/JCI74347
    1. Zhu E, Wang X, Zheng B, Wang Q, Hao J, Chen S, et al. miR-20b suppresses Th17 differentiation and the pathogenesis of experimental autoimmune encephalomyelitis by targeting RORγt and STAT3. J Immunol (2014) 192:5599–609.10.4049/jimmunol.1303488
    1. Hoy AM, Buck AH. Extracellular small RNAs: what, where, why? Biochem Soc Trans (2012) 40:886–90.10.1042/BST20120019
    1. Gandhi R, Healy B, Gholipour T, Egorova S, Musallam A, Hussain MS, et al. Circulating microRNAs as biomarkers for disease staging in multiple sclerosis. Ann Neurol (2013) 73:729–40.10.1002/ana.23880
    1. Søndergaard HB, Hesse D, Krakauer M, Sørensen PS, Sellebjerg F. Differential microRNA expression in blood in multiple sclerosis. Mult Scler (2013) 19:1849–57.10.1177/1352458513490542
    1. Siegel SR, Mackenzie J, Chaplin G, Jablonski NG, Griffiths L. Circulating microRNAs involved in multiple sclerosis. Mol Biol Rep (2012) 39:6219–25.10.1007/s11033-011-1441-7
    1. Farina NH, Wood ME, Perrapato SD, Francklyn CS, Stein GS, Stein JL, et al. Standardizing analysis of circulating microRNA: clinical and biological relevance. J Cell Biochem (2014) 115:805–11.10.1002/jcb.24745
    1. Keller A, Leidinger P, Steinmeyer F, Stähler C, Franke A, Hemmrich-Stanisak G, et al. Comprehensive analysis of microRNA profiles in multiple sclerosis including next-generation sequencing. Mult Scler (2014) 20:295–303.10.1177/1352458513496343
    1. Bergman P, James T, Kular L, Ruhrmann S, Kramarova T, Kvist A, et al. Next-generation sequencing identifies microRNAs that associate with pathogenic autoimmune neuroinflammation in rats. J Immunol (2013) 190:4066–75.10.4049/jimmunol.1200728
    1. Zhao S, Fung-Leung W-P, Bittner A, Ngo K, Liu X. Comparison of RNA-seq and microarray in transcriptome profiling of activated T cells. PLoS One (2014) 9:e78644.10.1371/journal.pone.0078644
    1. Xu X, Zhang Y, Williams J, Antoniou E, McCombie WR, Wu S, et al. Parallel comparison of Illumina RNA-seq and Affymetrix microarray platforms on transcriptomic profiles generated from 5-aza-deoxy-cytidine treated HT-29 colon cancer cells and simulated datasets. BMC Bioinformatics (2013) 14(Suppl 9):S1.10.1186/1471-2105-14-S9-S1
    1. Knowles DG, Röder M, Merkel A, Guigó R. Grape RNA-seq analysis pipeline environment. Bioinformatics (2013) 29:614–21.10.1093/bioinformatics/btt016
    1. Fenoglio C, Ridolfi E, Cantoni C, De Riz M, Bonsi R, Serpente M, et al. Decreased circulating miRNA levels in patients with primary progressive multiple sclerosis. Mult Scler (2013) 19:1938–42.10.1177/1352458513485654
    1. Cox MB, Cairns MJ, Gandhi KS, Carroll AP, Moscovis S, Stewart GJ, et al. microRNAs miR-17 and miR-20a inhibit T cell activation genes and are under-expressed in MS whole blood. PLoS One (2010) 5:e12132.10.1371/journal.pone.0012132
    1. Sievers C, Meira M, Hoffmann F, Fontoura P, Kappos L, Lindberg RLP. Altered microRNA expression in B lymphocytes in multiple sclerosis: towards a better understanding of treatment effects. Clin Immunol (2012) 144:70–9.10.1016/j.clim.2012.04.002
    1. Miyazaki Y, Li R, Rezk A, Misirliyan H, Moore C, Farooqi N, et al. A novel microRNA-132-surtuin-1 axis underlies aberrant B-cell cytokine regulation in patients with relapsing-remitting multiple sclerosis. PLoS One (2014) 9:e105421.10.1371/journal.pone.0105421
    1. Lindberg RLP, Hoffmann F, Mehling M, Kuhle J, Kappos L. Altered expression of miR-17-5p in CD4+ lymphocytes of relapsing-remitting multiple sclerosis patients. Eur J Immunol (2010) 40:888–98.10.1002/eji.200940032
    1. Lorenzi JCC, Brum DG, Zanette DL, de Paula Alves Souza A, Barbuzano FG, Dos Santos AC, et al. miR-15a and 16-1 are downregulated in CD4+ T cells of multiple sclerosis relapsing patients. Int J Neurosci (2012) 122:466–71.10.3109/00207454.2012.678444
    1. De Santis G, Ferracin M, Biondani A, Caniatti L, Rosaria Tola M, Castellazzi M, et al. Altered miRNA expression in T regulatory cells in course of multiple sclerosis. J Neuroimmunol (2010) 226:165–71.10.1016/j.jneuroim.2010.06.009
    1. Jernås M, Malmeström C, Axelsson M, Nookaew I, Wadenvik H, Lycke J, et al. microRNA regulate immune pathways in T-cells in multiple sclerosis (MS). BMC Immunol (2013) 14:32.10.1186/1471-2172-14-32
    1. Guerau-de-Arellano M, Smith KM, Godlewski J, Liu Y, Winger R, Lawler SE, et al. micro-RNA dysregulation in multiple sclerosis favours pro-inflammatory T-cell-mediated autoimmunity. Brain (2011) 134:3578–89.10.1093/brain/awr262
    1. Reijerkerk A, Lopez-Ramirez MA, van Het Hof B, Drexhage JAR, Kamphuis WW, Kooij G, et al. microRNAs regulate human brain endothelial cell-barrier function in inflammation: implications for multiple sclerosis. J Neurosci (2013) 33:6857–63.10.1523/JNEUROSCI.3965-12.2013
    1. Moore CS, Rao VTS, Durafourt BA, Bedell BJ, Ludwin SK, Bar-Or A, et al. miR-155 as a multiple sclerosis-relevant regulator of myeloid cell polarization. Ann Neurol (2013) 74:709–20.10.1002/ana.23967
    1. Noorbakhsh F, Ellestad KK, Maingat F, Warren KG, Han MH, Steinman L, et al. Impaired neurosteroid synthesis in multiple sclerosis. Brain (2011) 134:2703–21.10.1093/brain/awr200
    1. Dutta R, Chomyk AM, Chang A, Ribaudo MV, Deckard SA, Doud MK, et al. Hippocampal demyelination and memory dysfunction are associated with increased levels of the neuronal microRNA miR-124 and reduced AMPA receptors. Ann Neurol (2013) 73:637–45.10.1002/ana.23860
    1. Lu C, Huang X, Zhang X, Roensch K, Cao Q, Nakayama KI, et al. miR-221 and miR-155 regulate human dendritic cell development, apoptosis, and IL-12 production through targeting of p27kip1, KPC1, and SOCS-1. Blood (2011) 117:4293–303.10.1182/blood-2010-12-322503
    1. Grigoryev YA, Kurian SM, Hart T, Nakorchevsky AA, Chen C, Campbell D, et al. microRNA regulation of molecular networks mapped by global microRNA, mRNA, and protein expression in activated T lymphocytes. J Immunol (2011) 187:2233–43.10.4049/jimmunol.1101233
    1. György B, Szabó TG, Pásztói M, Pál Z, Misják P, Aradi B, et al. Membrane vesicles, current state-of-the-art: emerging role of extracellular vesicles. Cell Mol Life Sci (2011) 68:2667–88.10.1007/s00018-011-0689-3
    1. Pegtel DM, Cosmopoulos K, Thorley-Lawson DA, van Eijndhoven MAJ, Hopmans ES, Lindenberg JL, et al. Functional delivery of viral miRNAs via exosomes. Proc Natl Acad Sci U S A (2010) 107:6328–33.10.1073/pnas.0914843107
    1. Gibbings DJ, Ciaudo C, Erhardt M, Voinnet O. Multivesicular bodies associate with components of miRNA effector complexes and modulate miRNA activity. Nat Cell Biol (2009) 11:1143–9.10.1038/ncb1929
    1. Hunter MP, Ismail N, Zhang X, Aguda BD, Lee EJ, Yu L, et al. Detection of microRNA expression in human peripheral blood microvesicles. PLoS One (2008) 3:e3694.10.1371/journal.pone.0003694
    1. Zernecke A, Bidzhekov K, Noels H, Shagdarsuren E, Gan L, Denecke B, et al. Delivery of microRNA-126 by apoptotic bodies induces CXCL12-dependent vascular protection. Sci Signal (2009) 2:ra81–81.10.1126/scisignal.2000610
    1. Arroyo JD, Chevillet JR, Kroh EM, Ruf IK, Pritchard CC, Gibson DF, et al. Argonaute2 complexes carry a population of circulating microRNAs independent of vesicles in human plasma. Proc Natl Acad Sci U S A (2011) 108:5003–8.10.1073/pnas.1019055108
    1. Wang K, Zhang S, Weber J, Baxter D, Galas DJ. Export of microRNAs and microRNA-protective protein by mammalian cells. Nucleic Acids Res (2010) 38:7248–59.10.1093/nar/gkq601
    1. Vickers KC, Palmisano BT, Shoucri BM, Shamburek RD, Remaley AT. microRNAs are transported in plasma and delivered to recipient cells by high-density lipoproteins. Nat Cell Biol (2011) 13:423–33.10.1038/ncb2210
    1. Sáenz-Cuesta M, Irizar H, Castillo-Triviño T, Muñoz-Culla M, Osorio-Querejeta I, Prada A, et al. Circulating microparticles reflect treatment effects and clinical status in multiple sclerosis. Biomark Med (2014) 8:653–61.10.2217/bmm.14.9
    1. Sáenz-Cuesta M, Osorio-Querejeta I, Otaegui D. Extracellular vesicles in multiple sclerosis: what are they telling us? Front Cell Neurosci (2014) 8:100.10.3389/fncel.2014.00100
    1. Ridder K, Keller S, Dams M, Rupp A-K, Schlaudraff J, Turco DD, et al. Extracellular vesicle-mediated transfer of genetic information between the hematopoietic system and the brain in response to inflammation. PLoS Biol (2014) 12:e1001874.10.1371/journal.pbio.1001874
    1. Okoye IS, Coomes SM, Pelly VS, Czieso S, Papayannopoulos V, Tolmachova T, et al. microRNA-containing T-regulatory-cell-derived exosomes suppress pathogenic T helper 1 cells. Immunity (2014) 41:89–103.10.1016/j.immuni.2014.05.019
    1. Walker JD, Maier CL, Pober JS. Cytomegalovirus-infected human endothelial cells can stimulate allogeneic CD4+ memory T cells by releasing antigenic exosomes. J Immunol (2009) 182:1548–59.10.4049/jimmunol.182.3.1548
    1. Jy W, Minagar A, Jimenez JJ, Sheremata WA, Mauro LM, Horstman LL, et al. Endothelial microparticles (EMP) bind and activate monocytes: elevated EMP-monocyte conjugates in multiple sclerosis. Front Biosci (2004) 9:3137–44.10.2741/1466
    1. Hwang I, Shen X, Sprent J. Direct stimulation of naive T cells by membrane vesicles from antigen-presenting cells: distinct roles for CD54 and B7 molecules. Proc Natl Acad Sci U S A (2003) 100:6670–5.10.1073/pnas.1131852100
    1. Allan RS, Waithman J, Bedoui S, Jones CM, Villadangos JA, Zhan Y, et al. Migratory dendritic cells transfer antigen to a lymph node-resident dendritic cell population for efficient CTL priming. Immunity (2006) 25:153–62.10.1016/j.immuni.2006.04.017
    1. Pizzirani C, Ferrari D, Chiozzi P, Adinolfi E, Sandonà D, Savaglio E, et al. Stimulation of P2 receptors causes release of IL-1beta-loaded microvesicles from human dendritic cells. Blood (2007) 109:3856–64.10.1182/blood-2005-06-031377
    1. Blanchard N, Lankar D, Faure F, Regnault A, Dumont C, Raposo G, et al. Activation of human T cells induces the production of exosomes bearing the TCR/CD3/ζ complex. J Immunol (2002) 168:3235–41.10.4049/jimmunol.168.7.3235
    1. MacKenzie A, Wilson HL, Kiss-Toth E, Dower SK, North RA, Surprenant A. Rapid secretion of interleukin-1beta by microvesicle shedding. Immunity (2001) 15:825–35.10.1016/S1074-7613(01)00229-1
    1. Antonucci F, Turola E, Riganti L, Caleo M, Gabrielli M, Perrotta C, et al. Microvesicles released from microglia stimulate synaptic activity via enhanced sphingolipid metabolism. EMBO J (2012) 31:1231–40.10.1038/emboj.2011.489
    1. Bakhti M, Winter C, Simons M. Inhibition of myelin membrane sheath formation by oligodendrocyte-derived exosome-like vesicles. J Biol Chem (2011) 286:787–96.10.1074/jbc.M110.190009
    1. Basso M, Pozzi S, Tortarolo M, Fiordaliso F, Bisighini C, Pasetto L, et al. Mutant copper-zinc superoxide dismutase (SOD1) induces protein secretion pathway alterations and exosome release in astrocytes: implications for disease spreading and motor neuron pathology in amyotrophic lateral sclerosis. J Biol Chem (2013) 288:15699–711.10.1074/jbc.M112.425066
    1. Meinl E, Krumbholz M, Derfuss T, Junker A, Hohlfeld R. Compartmentalization of inflammation in the CNS: a major mechanism driving progressive multiple sclerosis. J Neurol Sci (2008) 274:42–4.10.1016/j.jns.2008.06.032
    1. Lovett-Racke AE, Yang Y, Racke MK. Th1 versus Th17: are T cell cytokines relevant in multiple sclerosis? Biochim Biophys Acta (2011) 1812:246–51.10.1016/j.bbadis.2010.05.012
    1. O’Connell RM, Kahn D, Gibson WSJ, Round JL, Scholz RL, Chaudhuri AA, et al. microRNA-155 promotes autoimmune inflammation by enhancing inflammatory T cell development. Immunity (2010) 33:607–19.10.1016/j.immuni.2010.09.009
    1. Brucklacher-Waldert V, Stuerner K, Kolster M, Wolthausen J, Tolosa E. Phenotypical and functional characterization of T helper 17 cells in multiple sclerosis. Brain (2009) 132:3329–41.10.1093/brain/awp289
    1. Polman CH, Reingold SC, Banwell B, Clanet M, Cohen JA, Filippi M, et al. Diagnostic criteria for multiple sclerosis: 2010 revisions to the McDonald criteria. Ann Neurol (2011) 69:292–302.10.1002/ana.22366
    1. Haghikia A, Haghikia A, Hellwig K, Baraniskin A, Holzmann A, Décard BF, et al. Regulated microRNAs in the CSF of patients with multiple sclerosis: a case-control study. Neurology (2012) 79:2166–70.10.1212/WNL.0b013e3182759621
    1. Swanton JK, Rovira A, Tintore M, Altmann DR, Barkhof F, Filippi M, et al. MRI criteria for multiple sclerosis in patients presenting with clinically isolated syndromes: a multicentre retrospective study. Lancet Neurol (2007) 6:677–86.10.1016/S1474-4422(07)70176-X
    1. Bitsch A, Brück W. Differentiation of multiple sclerosis subtypes: implications for treatment. CNS Drugs (2002) 16:405–18.10.2165/00023210-200216060-00004
    1. Waschbisch A, Atiya M, Linker RA, Potapov S, Schwab S, Derfuss T. Glatiramer acetate treatment normalizes deregulated microRNA expression in relapsing remitting multiple sclerosis. PLoS One (2011) 6:e24604.10.1371/journal.pone.0024604
    1. Meira M, Sievers C, Hoffmann F, Derfuss T, Kuhle J, Kappos L, et al. miR-126: a novel route for natalizumab action? Mult Scler (2014) 20:1363–70.10.1177/1352458514524998
    1. Meira M, Sievers C, Hoffmann F, Rasenack M, Kuhle J, Derfuss T, et al. Unraveling natalizumab effects on deregulated miR-17 expression in CD4+ T cells of patients with relapsing-remitting multiple sclerosis. J Immunol Res (2014) 2014:897249.10.1155/2014/897249

Source: PubMed

3
Abonnieren