Pathogenic Mechanisms Associated With Different Clinical Courses of Multiple Sclerosis

Hans Lassmann, Hans Lassmann

Abstract

In the majority of patients multiple sclerosis starts with a relapsing remitting course (RRMS), which may at later times transform into secondary progressive disease (SPMS). In a minority of patients the relapsing remitting disease is skipped and the patients show progression from the onset (primary progressive MS, PPMS). Evidence obtained so far indicate major differences between RRMS and progressive MS, but no essential differences between SPMS and PPMS, with the exception of a lower incidence in the global load of focal white matter lesions and in particular in the presence of classical active plaques in PPMS. We suggest that in MS patients two types of inflammation occur, which develop in parallel but partially independent from each other. The first is the focal bulk invasion of T- and B-lymphocytes with profound blood brain barrier leakage, which predominately affects the white matter, and which gives rise to classical active demyelinated plaques. The other type of inflammation is a slow accumulation of T-cells and B-cells in the absence of major blood brain barrier damage in the connective tissue spaces of the brain, such as the meninges and the large perivascular Virchow Robin spaces, where they may form aggregates or in most severe cases structures in part resembling tertiary lymph follicles. This type of inflammation is associated with the formation of subpial demyelinated lesions in the cerebral and cerebellar cortex, with slow expansion of pre-existing lesions in the white matter and with diffuse neurodegeneration in the normal appearing white or gray matter. The first type of inflammation dominates in acute and relapsing MS. The second type of inflammation is already present in early stages of MS, but gradually increases with disease duration and patient age. It is suggested that CD8+ T-lymphocytes remain in the brain and spinal cord as tissue resident cells, which may focally propagate neuroinflammation, when they re-encounter their cognate antigen. B-lymphocytes may propagate demyelination and neurodegeneration, most likely by producing soluble neurotoxic factors. Whether lymphocytes within the brain tissue of MS lesions have also regulatory functions is presently unknown. Key open questions in MS research are the identification of the target antigen recognized by tissue resident CD8+ T-cells and B-cells and the molecular nature of the soluble inflammatory mediators, which may trigger tissue damage.

Keywords: demyelination; inflammation; neurodegeneration; primary progressive MS; relapsing remitting MS; secondary progressive MS.

Figures

Figure 1
Figure 1
Active Lesions in early MS (acute and RRMS). (A) The dominant pathology in early MS is the presence of focal confluent demyelinated lesions in the white matter, many of them being in the stage of activity; section of a patient with acute multiple sclerosis, stained for myelin by immunohistochemistry for proteolipid protein. Magnification bar: 1 mm. (B) The classical active lesions in early MS develop around a central vein with inflammatory infiltrates, composed of CD8+ T-cells (red), CD20 positive B-cells (green), and few plasma cells (blue). While B-cells and plasma cells mainly remain in the perivascular space, the CD8+ T-cells also diffusely infiltrate the lesion parenchyme. The lesion (blue) is massively infiltrated by macrophages. Many of the lymphocytes are in the process of passing the vessel wall and this is associated with profound blood brain barrier leakage. This results in profound edema, which expands beyond the area of active demyelination (light blue). (C–E) Myelin staining (immunocytochemistry for proteolipid protein) shows patchy areas of active demyelination, which is associated with dense infiltration of the tissue by macrophages (D,E). (F, G) Immunohistochemistry for the T-cell marker CD8 shows perivascular accumulation of T-cells, and their diffuse infiltration of the lesion parenchyme. (H) The perivascular inflammatory infiltrates contain numerous CD20+ B-lymphocytes. (I,J) Staining for IgG reveals massive leakage of the blood brain barrier and only a small number of IgG containing plasma cells in the perivascular space (I); the profound blood brain barrier leakage is also reflected by extensive leakage of fibrinogen through the inflamed vessels (J). (K) A subset of macrophages expresses the activation marker CD163, a feature which is typically found in active MS lesions. The magnification bars in the figures (C,G,I) represent 100 μm. Similar histological images as shown in this figure have been previously published. Structure of the lesions: Frischer et al. (46); Inflammatory reaction: Frischer et al. (47); Machado Santos et al. (37); Microglia and macrophages: Zrzavy et al. (38); Fibrin and blood brain barrier injury: Hochmeister et al. (48).
Figure 2
Figure 2
Inflammatory reaction in the brain of patients with progressive MS and its relation to active demyelination and neurodegeneration. The inflammatory reaction in the brain of patients with progressive MS is mainly seen in the large connective tissue spaces of the meninges and the periventricular Virchow Robin spaces. These inflammatory sites mainly contain CD8+ T-cells, a major component of CD20+ B-cells and a variable number of plasma cells and may in their most severe manifestations become organized in structures with features of tertiary lymph follicles (green dots). In addition there are perivascular cuffs mainly composed of CD8+ T-cells, which are more broadly dispersed within the white matter of the brain (red dots). Inflammation with T-cells, B-cells and Plasma cells (green dots) is associated with slow expansion of demyelinated lesions, defined by a rim of activated microglia cells, which in part contain early myelin degradation products in the cortex and the white matter (thick blue lines). Active demyelination and diffuse tissue injury occurs at a distance from the lymphocytic infiltrates and may, thus, be propagated by a soluble demyelinating or neurotoxic factor. Inactive plaques (thin green lines) can still be centered by a vein with a dominant infiltrate by CD8+ T-cells (red dots).
Figure 3
Figure 3
Slowly expanding lesions in the progressive stage of MS in the cortex and the white matter. (A) Active cortical lesions are associated with inflammatory infiltrates in the meninges, which are composed of CD8+ T-cells (red), CD20+ B-cells (green) and plasma cells (blue). Active demyelination occurs at a distance of the inflammatory infiltrates and is associated with activated microglia (blue lesion rim). The lesions gradually expand from the pial surface of the cortex toward the depth of the gray matter. Lymphocyte infiltrates are rare or completely absent in the cortical tissue and in particular at the zone of active demyelination. It is suggested that the inflammatory infiltrates in the meninges produce a soluble factor, which induces demyelination and neurodegeneration either directly or indirectly through microglia activation (arrows). (B) In slowly expanding lesions in the white matter T-cell, B-cell and plasma cell infiltrates are present in the large perivascular Virchow Robin spaces. Active demyelination and neurodegeneration occurs at a distance and is associated with microglia activation. Also in these lesions it is suggested that demyelination and neurodegeneration is driven by a soluble factor, produced by the perivascular lymphocytes or plasma cells (arrows). (C–F) Active cortical lesion in a patient with progressive MS. Subpial myelin is completely lost in an area with meningeal inflammation (C); CD8+ T-cells are present in the meningeal infiltrates, but do not enter the cortical parenchyme (D); The meningeal infiltrates also contain IgG positive plasma cells (E), there is however no indication of IgG leakage from the vessels into the tissue, suggesting an intact blood brain barrier. Activated microglia and macrophages are seen at the site of active demyelination in the depth of the gray matter (F). (G,H) Slowly expanding lesion in the white matter of a patient with progressive MS. The inactive plaque center contains vessels with perivascular cuffs of lymphocytes but the active demyelination at the lesion edge is associated with a rim of activated microglia (G,H). Lymphocytes, such as for instance CD8+ T-cells and B-cells are present in the large perivascular space of the vessels, but there is little or no infiltration into the lesion parenchyme (I,J). No fibrinogen leakage is observed around inflamed vessels, indicating intact blood brain barrier function (K). Magnification bar representative for all images: 100 μm. Similar histological images as shown in this figure have been previously published. Structure of the lesions: Frischer et al. (46); Inflammatory reaction: Frischer et al. (47); Machado Santos et al. (37); Microglia and macrophages: Zrzavy et al. (38); Fibrin and blood brain barrier injury: Hochmeister et al. (48).

References

    1. Lassmann H, Brück W, Lucchinetti C. The immunopathology of multiple sclerosis: an overview. Brain Pathol. (2007) 17:210–8. 10.1111/j.1750-3639.2007.00064.x
    1. Lublin FD, Reingold SC, Cohen JA, Cutter GR, Sørensen PS, Thompson AJ, et al. . Defining the clinical course of multiple sclerosis. The 2013 revision. Neurology (2014) 83:1–9. 10.1212/WNL.0000000000000560
    1. Trapp BD, Nave KA. Multiple sclerosis: an immune or neurodegenerative disorder? Ann Rev Neurosci. (2008) 31:247–2696. 10.1146/annurev.neuro.30.051606.094313
    1. Antel J, Antel S, Caramanos Z, Arnold DL, Kuhlmann T. Promary progressive multiple sclerosis: part of the MS disease spectrum or separate disease entity?- Acta Neuropathol. (2012) 123:627–38. 10.1007/s00401-012-0953-0
    1. Mahad DH, Trapp BD, Lassmann H. Pathological mechanisms in progressive multiple sclerosis. Lancet Neurol. (2015) 14:183–93. 10.1016/S1474-4422(14)70256-X
    1. Hohlfeld R, Dornmair K, Meinl E, Wekerle H. The search for the target antigens of multiple sclerosis, part 1: autoreactive CD4+ T lymphocytes as pathogenic effectors and therapeutic targets. Lancet Neurol. (2016) 15:198–209. 10.1016/S1474-4422(15)00334-8
    1. Hohlfeld R, Dornmair K, Meinl E, Wekerle H. The search for the target antigens of multiple sclerosis, part 2: CD8+ T cells, B cells, and antibodies in the focus of reverse-translational research. Lancet Neurol. (2016) 15:317–31. 10.1016/S1474-4422(15)00313-0
    1. Claes N, Fraussen J, Stinissen P, Hupperts R, Somers V. B cells are multifunctional players in multiple sclerosis pathogenesis: insights from therapeutic interventions. Front Immunol. (2015) 6:642. 10.3389/fimmu.2015.00642
    1. Dargahi N, Katsara M, Tselios T, Androutsou ME, de Courten M, Matsoukas J, et al. . Multiple sclerosis: immunopathology and treatment update. Brain Sci. (2017) 7:E78. 10.3390/brainsci7070078
    1. Rice CM, Cottrell D, Wilkins A, Scolding NJ. Primary progressive multiple sclerosis: progress and chalenges. J Neurol Neurosurg Psychiat. (2013) 84:1100–6. 10.1136/jnnp-2012-304140
    1. Abdelhack A, Weber MS, Tumani H. Primary progressive multiple sclerosis: putting together the puzzle. Front Neurol. (2017) 8:234 10.3389/fneur.2017.00234
    1. Confafreux C, Vukusic S, Moreau T, Adeleine P. Relapses and progression of disability in multiple sclerosis. N Engl J Med. (2000) 343:1430–8. 10.1056/NEJM200011163432001
    1. Leray E, Yaouang J, Le Page E, Coustans M, Laplaud D, Oger J, et al. . Evidence of a two stage disability progression in multiple sclerosis. Brain (2010) 133:1900–13. 10.1093/brain/awq076
    1. Kuchling J, Ramien C, Bozin I, Dörr J, Harms L, Rosche B, et al. Identical lesions morphology in primary progressive and relapsing-remitting MS. An ultrahigh filed MRI study. Mult Scler. (2014) 20:1866–71. 10.1177/1352458514531084
    1. Scalfari A, Lederer C, Daumer M, Nicholas R, Ebers GC, Muraro PA. The relationship of age with the clinical phenotype in multiple sclerosis. Mult Scler. (2016) 22:1750–8. 10.1177/1352458516630396
    1. Zeydan B, Kantarci OH. Progressive forms of multiple sclerosis: distinct entity of age dependent phenomena. Neurol Clin. (2018) 36:163–72. 10.1016/j.ncl.2017.08.006
    1. Ebers GC. Natural history of primary progressive multiple sclerosis. Mult Scler. (2004) 10(Suppl. 1):S8–13. 10.1191/1352458504ms1025oa
    1. Sadovnick AD. Differential effects of genetic susceptibility factors in males and females with multiple sclerosis. Clin Immunol. (2013) 149:170–5. 10.1016/j.clim.2013.05.002
    1. International Multiple Sclerosis Genetcis Consortium Analysis of immune-related loci identifies 48 new susceptibility variants for multiple sclerosis. Nat. Genet. (2013) 45:1353–60. 10.1038/ng.2770
    1. International Multiple Sclerosis Genetcis Consortium Low-frequency abd rare-coding variation contributes to multiple sclerosis risk. Cell (2018) 175:1679–87.e7. 10.1016/j.cell.2018.09.049
    1. Chataway J, Mander A, Robertson N, Sawcer S, Deans J, Fraser M, et al. . Multiple sclerosis in sibling pairs: an analysis of 250 families. J Neurol Neurosurg Psychiatr. (2001) 71:757–61. 10.1136/jnnp.71.6.757
    1. Oturai AB, Ryder LP, Fredrickson S, Myhr KM, Celius EG, Harbo HF, et al. Concordance of disease course and age of onset in Scandinavian multiple sclerosis co-affected sib pairs. Mult Scler. (2004) 10:5–8. 10.1191/1352458504ms975oa
    1. Jia X, Madireddy L, Caillier S, Santaniello A, Esposito F, Comi G, et al. Genomic sequencing uncovers phenocopies in primary progressive multiple sclerosis. Ann Neurol. (2017) 84:51–63. 10.1002/ana.25263
    1. Pan G, Simpson S, van der Mei I, Charlesworth JC, Lucas R, Ponsonby AL, et al. . Role of genetic susceptibility variants in predicting clinical course in multiple sclerosis: a cohort study. J Neuro Neurosurg Psychiat. (2016) 87:1204–11. 10.1136/jnnp-2016-313722
    1. Wang Z, Sadovnick AD, Traboulsee AL, Ross JP, Bernales CQ, Encarnacion M, et al. . Nuclear receptor NR1H3 in familial multiple sclerosis. Neuron (2016) 90:948–54. 10.1016/j.neuron.2016.04.039
    1. International Multiple Sclerosis Genetcis Consortium. NR1H3 p.Arg415Gln is not associated to multiple sclerosis risk. Neuron(2016) 92:333–5. 10.1016/j.neuron.2016.09.052
    1. Sawcer S, Hellenthal G, Pirinen M, Spencer CC, Patsopoulos NA, Moutsianas L, et al. . Genetic risk and a primary role for cell-mediated immune mechanisms in multiple sclerosis. Nature (2011) 476:214–9. 10.1038/nature10251
    1. Housley WJ, Pitt D, Hafler DA. Biomarkers in multiple sclerosis. Clin Immunol. (2015) 161:51–8. 10.1016/j.clim.2015.06.015
    1. Paul A, Comabella M, Gandhi R. Biomarkers in multiple sclerosis. Cold Spring Harb Perspect Med. (2018) a029058 10.1101/cshperspect.a029058
    1. Abdelhak A, Hottenrott T, Mayer C, Hintereder G, Zettl U, Stich O, et al. CSF profile in primary progressive multiple sclerosis; reexploring the basics. PLoS ONE (2017) 12:e0182647 10.1371/journal.pone.0182647
    1. Thompson AJ, Reingold SC, Cohen JA. Applying the 2017 McDonald diagnostic criteria for multiple sclerosis - Authors' reply. Lancet Neurol. (2018) 17:499–500. 10.1016/S1474-4422(18)30168-6
    1. Hohlfeld R. Immunologic factors in primary progressive multiple sclerosis. Mult Scler. (2004) 10:S16–22. 10.1191/1352458504ms1026oa
    1. Bielekova B, Komori M, Xu Q, Reich DS, Wu T. Cerebrospinal fluid Il12p40, CXCL13 and Il-8 as a combinatorial biomarker of active intrathecal inflammation. PLoS ONE (2012) 7:e48370 10.1371/journal.pone.0048370
    1. Han S, Lin YC, Wu T, Salgado AD, Mexhitaj I, Wuest SC, et al. Comprehensive immunophenotyping of cerebrospinal fluid cells in patients with neuroimmunological disease. J Immunol. (2014) 192:2551–63. 10.4049/jimmunol.1302884
    1. Barbour C, Kosa P, Komori M, Tanigawa M, Masvekar R, Wu T, et al. Molecular-based diagnosis of multiple sclerosis and ist progressive stage. Ann Neurol. (2017) 82:795–812. 10.1002/ana.25083
    1. Wurth S, Kuenz B, Bsteh G, Ehling R, DiPauli F, Heegen H, et al. . Cerebrospinal fluid B cells and disease progression in multiple sclerosis - a longitudinal study. PLoS ONE (2017) 12:e0182462. 10.1371/journal.pone.0182462
    1. Machado-Santos J, Saji E, Tröscher A, Paunovic M, Liblau R, Gabriely G, et al. . The compartmentalized inflammatory response in the multiple sclerosis brain is composed of tissue-resident CD8+ T lymphocytes and B cells. Brain (2018) 141:2066–82. 10.1093/brain/awy151
    1. Zrzavy T, Hametner S, Wimmer I, Butovsky O, Weiner H, Lassmann H. Loss of ‘homeostatic' microglia and patterns of their activation in active multiple sclerosis. Brain (2017) 140:1900–13. 10.1093/brain/awx113
    1. Thompson EJ, Keir G. Laboratory investigation of cerebrospinal fluid proteins. Ann Clin Biochem. (1990) 27:425–35. 10.1177/000456329002700503
    1. Cepok S, Zhou D, Vogel F, Rosche B, Grummel V, Sommer N, et al. . The immune response at onset and during recovery from Borrelia buf'rgdorferi meningoradiculitis. Arch Neurol. (2003) 60:849–55. 10.1001/archneur.60.6.849
    1. Cepok S, Rosche B, Brummel V, Vogel F, Zhou D, Sayn J, et al. . Short-lived plasma blasts are the main B-cell effector subset during the course of multiple sclerosis. Brain (2005) 128:1667–76. 10.1093/brain/awh486
    1. Magliozzi R, Howell OW, Nicholas R, Cruciani C, Castellaro M, Romualdi C, et al. . Inflammatory intrathecal profiles and cortical damage in multiple sclerosis. Ann Neurol. (2018) 83:739–55. 10.1002/ana.25197
    1. Mane-Martinez MA, Olsson B, Bau L, Matas E, Cobo-Calvo A, Andreasson U, et al. . Glial and neuronal markers in cerebrospinal fluid in different types of multiple sclerosis. J Neuroimmunol. (2016) 299:112–7. [Epub ahead of print] 10.1016/j.jneuroim.2016.08.004
    1. Barro C, Benkert P, Disanto G, Tsagkas C, Amann M, Naegelin Y, et al. Serum neurofilament as a predictor of disease worsening and brain and spinal cord atrophy in multiple sclerosis. Brain (2018) 141:2382–91. 10.1093/brain/awy154
    1. Stoessel D, Stellmann JP, Willimg A, Behrens B, Rosenkranz SC, Hodecker SC, et al. . Metabolomic profiles for primary progressive multiple sclerosis stratification and disease course monitoring. Front Hum Neurosci. (2018) 12:226. 10.3389/fnhum.2018.00226
    1. Frischer JM, Weigand SD, Guo Y, Kale N, Parisi JE, Pirko I, et al. . Clinical and pathological insights into the dynamic nature of the white matter multiple sclerosis plaque. Ann Neurol. (2015) 78:710–21. 10.1002/ana.24497
    1. Frischer JM, Bramow S, Dal Bianco A, Lucchinetti CF, Rauschka H, Schmidbauer M, et al. . The relation between inflammation and neurodegeneration in multiple sclerosis brains. Brain (2009) 132:1175–89. 10.1093/brain/awp070
    1. Hochmeister S, Grundtner R, Bauer J, Engelhardt B, Lyck R, Gordon G, et al. . Dysferlin is a new marker for leaky brain blood vessels in multiple sclerosis. J Neuropathol Exp Neurol. (2006) 65:855–65. 10.1097/01.jnen.0000235119.52311.16
    1. Kutzelnigg A, Lucchinetti CF, Stadelmann C, Bruck W, Rauschka H, Bergmann M, et al. . Cortical demyelination and diffuse white matter injury in multiple sclerosis. Brain (2005) 128(Pt 11):2705–12. 10.1093/brain/awh641
    1. Haider L, Simeonidou C, Steinberger G, Hametner S, Grigoriadis N, Deretzi G, et al. . Multiple sclerosis deep grey matter: the relation between demyelination, neurodegeneration, inflammation and iron. J Neurol Neurosurg Psychiatry (2014) 85:1386–95. 10.1136/jnnp-2014-307712
    1. Schmierer K, Miquel ME. Magnetic resonance imaging correlates of neuro-axonal pathology in the MS spinal cord. Brain Pathol. (2018) 28:765–72. 10.1111/bpa.12648
    1. Bjartmar C, Kidd G, Mork S, Rudick R, Trapp BD. Neurological disability correlates with spinal cord axonal loss and reduced N-acetyl aspartate in chronic multiple sclerosis patients. Ann-Neurol. (2000) 48:893–901. 10.1002/1531-8249(200012)48:6<893::AID-ANA10>;2-B
    1. Patrikios P, Stadelmann C, Kutzelnigg A, Rauschka H, Schmidbauer M, Laursen H, et al. . Remyelination is extensive in a subset of multiple sclerosis patients. Brain (2006) 129:3165–72. 10.1093/brain/awl217
    1. Patani R, Balaratnam M, Vora A, Reynolds R. Remyelination can be extensive in multiple sclerosis despite a long disease course. Neuropath Appl Neurobiol. (2007) 33:277–87. 10.1111/j.1365-2990.2007.00805.x
    1. Hayashi T, Morimoto C, Burks JS, Kerr C, Hauser SL. Dual-label immunocytochemistry of the active multiple sclerosis lesion: major histocompatibility complex and activation antigens. Ann Neurol. (1988) 24:523–31. 10.1002/ana.410240408
    1. Booss J, Esiri MM, Tourtellotte WW, Mason DY. Immunohistological analysis of T lymphocyte subsets in the central nervous system in chronic progressive multiple sclerosis. J Neurol Sci. (1983) 62:219–32. 10.1016/0022-510X(83)90201-0
    1. van Nierop GP, van Luijn MM, Michels SS, Melief MJ, Janssen M, Langerak AW, et al. Phenotypic and functional characterization of T cells in white matter lesions of multiple sclerosis patients. Acta Neuropathol. (2017) 134:383–401. 10.1007/s00401-017-1744-4
    1. Hauser SL, Waubant E, Arnold DL, Vollmer T, Antel J, Fox RJ, et al. . B-cell depletion with rituximab in relapsing-remitting multiple sclerosis. N Engl J Med. (2008) 358:676–88. 10.1056/NEJMoa0706383
    1. Montalban X, Hauser SL, Kappos L, Arnold DL, Bar-Or A, Comi G, et al. . Ocrelizumab versus placebo in primary progressive multiple sclerosis. N Engl J Med. (2017) 376:209–20. 10.1056/NEJMoa1606468
    1. Lisak RP, Benjamins JA, Nedelkoska L, Barger JL, Ragheb S, Fan B, et al. . Secretory products of multiple sclerosis B cells are cytotoxic to oligodendroglia in vitro. J Neuroimmunol. (2012) 246:85–95. 10.1016/j.jneuroim.2012.02.015
    1. Lisak RP, Nedelkoska L, Benjamins JA, Schalk D, Bealmear B, Touil H, et al. . B cells from patients with multiple sclerosis induce cell death via apoptosis in neurons in vitro. J Neuroimmunol. (2017) 309:88–99. 10.1016/j.jneuroim.2017.05.004
    1. Revesz T, Kidd D, Thompson AJ, Barnard RO, McDonals WI. A comparison of the pathology of primary and secondary progressive multiple sclerosis. Brain (1994) 117:759–65. 10.1093/brain/117.4.759
    1. Esiri MM, Gay D. Immmunological and neuropathological significance of the Virchow-Robin space. J Neurol Sci. (1990) 100:3–8. 10.1016/0022-510X(90)90004-7
    1. Serafini B, Rosicarelli B, Magliozzi R, Stigliano E, Aloisi F. Detection of ectopic B-cell follicles with germinal centers in the meninges of patients with secondary progressive multiple sclerosis. Brain Pathol. (2004) 14:164–74. 10.1111/j.1750-3639.2004.tb00049.x
    1. Howell OW, Reeves CA, Nicholas R, Carassiti D, Radotra B, Gentleman SM, et al. . Meningeal inflammation is widespread and linked to cortical pathology in multiple sclerosis. Brain (2011) 134(Pt 9):2755–71. 10.1093/brain/awr182
    1. Choi S, Howell OW, Carassiti D, Magliozzi R, Gveric D, Muraro PA, et al. . Meningeal inflammation plays a role in the pathology of primary progressive multiple sclerosis. Brain (2012) 135:2925–37. 10.1093/brain/aws189
    1. Haider L, Zrzavy T, Hametner S, Höftberger R, Bagnato F, Grabner G, et al. . The topograpy of demyelination and neurodegeneration in the multiple sclerosis brain. Brain (2016) 139:807–15. 10.1093/brain/awv398
    1. Kuhlmann T, Ludwin S, Prat A, Antel J, Bruck W, Lassmann H. An updated histological classification system for multiple sclerosis lesions. Acta Neuropathol. (2017) 133:13–24. 10.1007/s00401-016-1653-y
    1. Luchetti S, Fransen NL, van Eden CG, Ramaglia V, Mason M, Huitinga I. Progressive multiple sclerosis patients show substantial lesion activity that correlates with clinical disease severity and sex. A retrospective autopsy cohort analysis. Acta Neuropathol. (2018) 135:511–28. 10.1007/s00401-018-1818-y
    1. Dal-Bianco A, Grabner G, Kronnerwetter C, Weber M, Höftberger R, Berger T, et al. . Slow expansion of multiple sclerosis iron rim lesions: pathology and 7 T magnetic resonance imaging. Acta Neuropathol. (2017) 133:25–42. 10.1007/s00401-016-1636-z
    1. Thompson AJ, Kermode AG, Wicks D, MacManus DG, Kendall BE, Kingsley DP, et al. . Major differences in the dynamics of primary and secondary progressive multiple sclerosis. Ann Neurol. (1991) 29:53–62. 10.1002/ana.410290111
    1. Thompson AJ, Polamn CH, Miller DH, McDonals WI, Brochet B, Filippi M, et al. . Primary progressive multiple sclerosis. Brain (1997) 120:1085–96. 10.1093/brain/120.6.1085
    1. Bramow S, Frischer JM, Lassmann H, Koch-Henriksen N, Lucchinetti CF, Sørensen PS, et al. . Demyelination versus remyelination in progressive multiple sclerosis. Brain (2010) 133:2983–98. 10.1093/brain/awq250
    1. Trapp DB, Vignos M, Dudman J, Chang A, Fisher E, Staugaitis SM, et al. . Cortical neuronal densities and cerebral white matter demyelination in multiple sclerosis: a retrospective study. Lancet Neurol. (2018) 17:870–84. 10.1016/S1474-4422(18)30245-X
    1. Zarei M, Chandran S, Compston A, Hodges J. Cognitive presentation of multiple sclerosis: evidence for a cortical variant. J Neurol Neurosurg Psychiatr. (2003) 74:872–7. 10.1136/jnnp.74.7.872
    1. Kutzelnigg A, Faber-Rod JC, Bauer J, Lucchinetti CF, Sorensen PS, Laursen H, et al. . Widespread demyelination in the cerebellar cortex in multiple sclerosis. Brain Pathol. (2007) 17:38–44. 10.1111/j.1750-3639.2006.00041.x
    1. Howell OW, Schulz-Trieglaff EK, Carassiti D, Gentleman SM, Nicholas R, Roncaroli F, et al. . Extensive grey matter pathology in the cerebellum in multiple sclerosis is linked to inflammation in the subarachnoid space. Neuropath Appl Neurobiol. (2015) 41:798–813. 10.1111/nan.12199
    1. Geurts JJ, Bo L, Roosendaal SD, Hazes T, Daniels R, Barkhof F, et al. . Extensive hippocampal demyelination in multiple sclerosis. J Neuropath Exp Neurol. (2007) 66:819–27. 10.1097/nen.0b013e3181461f54
    1. Pitt D, Boster A, Pei W, Wohleb E, Jasne A, Zachariah CR, et al. . Imaging cortical lesions in multiple sclerosis with ultra-high-filed magnetic resonance imaging. Arch Neurol. (2010) 67:812–8. 10.1001/archneurol.2010.148
    1. Schmierer K, Parkes HG, So PW, So PW, Brandner A, Ordidge RJ, et al. . High field (9.4 Tesla) magnetic resonance imaging of cortical grey matter lesions in multiple sclerosis. Brain (2010) 133:858–67. 10.1093/brain/awp335
    1. Hametner S, Bianco AD, Trattnig S, Lassmann H. Iron related changes in MS lesions and their validity to characterize MS lesion types and dynamics with ultra-high filed magnetic resonance imaging. Brain Pathol. (2018) 28:743–9. 10.1111/bpa.12643
    1. Lucchinetti CF, Popescu BF, Bunyan RF, Moll NM, Roemer SF, Lassmann H, et al. . Inflammatory cortical demyelination in early multiple sclerosis. N Engl J Med. (2011) 365:2188–97. 10.1056/NEJMoa1100648
    1. Magliozzi R, Howell O, Vora A, et al. . Meningeal B-cell follicles in secondary progressive multiple sclerosis associate with early onset of disease and severe cortical pathology. Brain (2007) 130:1089–104. 10.1093/brain/awm038
    1. Magliozzi R, Howell OW, Reeves C, Roncaroli F, Nicholas R, Serafini B, et al. . A Gradient of neuronal loss and meningeal inflammation in multiple sclerosis. Ann Neurol. (2010) 68:477–93. 10.1002/ana.22230
    1. Kutzelnigg A, Lassmann H. Cortical demyelination in multiple sclerosis: a substrate for cognitive deficits ? J Neurol Sci. (2006) 245:123–6. 10.1016/j.jns.2005.09.021
    1. Bornstein MB, Appel SH. Tissue culture studies of demyelination. Ann N Y Acad Sci. (1965) 122:280–6. 10.1111/j.1749-6632.1965.tb20212.x
    1. Vidaurre OG, Haines JD, Katz Sand I, Adula KP, Huynh JL, McGraw CA, et al. Cerebrospinal fluid ceramides from patents with multiple sclerosis impair neuronal bioenergetics. Brain (2014) 137:2271–86. 10.1093/brain/awu139
    1. Chiou B, Lucassen E, Sather M, Kallianpur A, Connor J. Semaphorin 4A and H-Ferritin utilize Tim-1 on human oligodendrocytes: a novel neuro-immune axis. Glia (2018) 66:1317–30. 10.1002/glia.23313
    1. Fischer MT, Wimmer I, Hoftberger R, Gerlach S, Haider L, Zrzavy T, et al. . Disease-specific molecular events in cortical multiple sclerosis lesions. Brain (2013) 136:1799–815. 10.1093/brain/awt110
    1. Vercellino M, Masera S, Lorenzatti M, Condello C, Merola A, Mattioda A, et al. Demyelination, inflammation, and neurodegeneration in multiple sclerosis deep grey matter. J Neuropath Exp Neurol. (2009) 68:489–502. 10.1097/NEN.0b013e3181a19a5a
    1. Huitinga I, De Groot CJ, van der Valk P, Kamphorst W, Tilders FJ, Swaab DF. Hypothalamic lesions in multiple sclerosis. J Neuropath Exp Neurol. (2001) 60:1208–18. 10.1093/jnen/60.12.1208
    1. Mews I, Bergmann M, Bunkowski S, Gullotta F, Brück W. Oligodendrocyte and axon pathology in clinically silent multiple sclerosis lesions. Mult Scler. (1998) 4:55–62. 10.1177/135245859800400203
    1. Kornek B, Storch M, Weissert R, Wallstroem E, Stefferl A, Olsson T, et al. . Multiple sclerosis and chronic autoimmune encephalomyelitis: a comparative quantitative study of axonal injury in active, inactive and remyelinated lesions. Amer J Pathol. (2000) 157:267–76. 10.1016/S0002-9440(10)64537-3
    1. Singh S, Dallenga T, Winkler A, Roemer S, Maruschak B, Siebert H, et al. . Relationship of acute axonal damage, Wallerian degeneration and clinical disability in multiple sclerosis. J Neurinflammation (2017) 14:57. 10.1186/s12974-017-0831-8
    1. Androdias G, Reynolds R, Chanal M, Ritleng C, Confavreux C, Nataf S. Meningeal T cells associate with diffuse axonal loss in multiple sclerosis spinal cords. Ann Neurol. (2010) 68:465–76. 10.1002/ana.22054
    1. Moore GR, Laule C, Mackay A, Leung E, Li DK, Zhao G, et al. . Dirty-appearing white matter in multiple sclerosis: preliminary observations of myelin phospholipid and axonal loss. J Neurol. (2008) 255:1802–11. 10.1007/s00415-008-0002-z
    1. Laule C, Vavasour IM, Leung E, Li DK, Kozlowski P, Traboulsee AL, et al. . Pathological basis of diffusely abnormal white matter: insights from magnetic resonance imaging and histology. Mult Scler. (2011) 17:144–50. 10.1177/1352458510384008
    1. Laule C, Pavlova V, Leung E, Zhao G, MacKay AL, Kozlowski P, et al. . Diffusely abnormal white matter in multiple sclerosis; further histologic studies provide evidence for a primary lipid abnormality with neurodegeneration. J Neuropath Exp Neurol. (2013) 72:42–52. 10.1097/NEN.0b013e31827bced3
    1. Lassmann H, Bradl M. Multiple sclerosis: experimental models and reality. Acta Neuropathol. (2017) 133:223–44. 10.1007/s00401-016-1631-4
    1. Smolders J, Heutinck KM, Fransen NL, Remmerswaal EBM, Hombrink P, Ten Berge IJM, et al. . Tissue-resident memory T cells populate the human brain. Nat Commun. (2018) 9:4593. 10.1038/s41467-018-07053-9
    1. Jarius S, Eichhorn P, Franciotta D, Petereit HF, Akman-Demir G, Wich M, et al. The MRZ reaction as a highly specific marker of multiple sclerosis: re-evaluation and structures review of the literature. J Neurol. (2017) 264:453–66. 10.1007/s00415-016-8360-4
    1. Babbe H, Roers A, Waisman A, Lassmann H, Goebels N, Hohlfeld R, et al. . Clonal expansions of CD8(+) T cells dominate the T cell infiltrate in active multiple sclerosis lesions as shown by micromanipulation and single cell polymerase chain reaction. J Exp Med. (2000) 192:393–404. 10.1084/jem.192.3.393
    1. Sinha S, Boyden AW, Itani FR, Crawford MP, Karandikar NJ. CD8(+) T-cells as immune regulators pf multiple sclerosis. Front Immunol. (2015) 6:619 10.3389/fimmu.2015.00619
    1. Boyden AW, Brate AA, Karandikar NJ. Early IFNg-mediated and late perforin mediated suppression of pathogenic CD4 T cell responses are both required for inhibition of demyelinating disease by CNS-specific autoregulatory CD8 cells. Front Immunol. (2018) 9:2336 10.3389/fimmu.2018.02336
    1. Lehmann-Horn K, Kinzel S, Weber MS. Deciphering the role of B cells in multiple sclerosis. Towards specific targeting of pathogenic functions. Int J Mol Sci. (2017) 18:E2048. 10.3390/ijms18102048
    1. Li R, Patterson KR, Bar-Or A. Reassessing B cell contributions in multiple sclerosis. Nat Immunol. (2018) 19:696–707. 10.1038/s41590-018-0135-x
    1. Lucchinetti C, Brück W, Parisi J, Scheithauer B, Rodriguez M, Lassmann H. Heterogeneity of multiple sclerosis lesions: implications for the pathogenesis of demyelination. Ann Neurol. (2000) 47:707–17. 10.1002/1531-8249(200006)47:6<707::AID-ANA3>;2-Q
    1. Jarius S, Ruprecht K, Kleiter I, Borisow N, Asgari N, Pitarokoili K, et al. . MOG-IgG in NMO and related disorders: a multicenter study of 50 patients. Part 2: Epidemiology, clinical presentation, radiological and laboratory features, treatment responses, and long-term outcome. J Neuroinflammation (2016) 13:280. 10.1186/s12974-016-0718-0
    1. Lily O, Palace J, Vincent A. Serum antibodies to sell surface determinants in multiple sclerosis: a flow cytometric study. Brain (2004) 127:269–79. 10.1093/brain/awh031
    1. Blauth K, Soltys J, Matschulat A, Reiter CR, Ritchie A, Baird NL, et al. . Antibodies produced by clonally expanded plasma cells in multiple sclerosis cerebrospinal fluid cause demyelination of spinal cord explants. Acta Neuropathol. (2015) 130:765–81. 10.1007/s00401-015-1500-6
    1. Aloisi F, Pujol-Borrell R. Lymphoid neogenesis in chronic inflammatory diseases. Nat Rev Immunol. (2006) 6:205–17. 10.1038/nri1786
    1. Schmied M, Breitschopf H, Gold R, Zischler H, Rothe G, Wekerle H, et al. . Apoptosis of T lymphocytes in experimental autoimmune encephalomyelitis. Evidence for programmed cell death as a mechanism to control inflammation in the brain. Am J Pathol. (1993) 143:446–52.
    1. Lassmann H, van Horssen J, Mahad D. Progressive multiple sclerosis: pathology and pathogenesis. Nat Rev Neurol. (2012) 8:647–56. 10.1038/nrneurol.2012.168
    1. Schenkel JM, Masopust D. Tissue-resident memory T cells. Immunity (2014) 41:886–97. 10.1016/j.immuni.2014.12.007
    1. Steinbach K, Vincenti I, Kreutzfeldt M, Page N, Muschaweckh A, Wagner I, et al. . Brain-resident memory T cells represent an autonomous cytotoxic barrier to viral infection. J Exp Med. (2016) 213:1571–87. 10.1084/jem.20151916
    1. Hauser SL. The Charcot Lecture / Beating MS: a story of B cells, with twists and turns. Mult Scler J. (2015) 21:8–21. 10.1177/1352458514561911
    1. Storch MK, Bauer J, Linington C, Olsson T, Weissert R, Lassmann H. Cortical demyelination can be modeled in specific rat models of autoimmune encephalomyelitis and is major histocompatibility complex (MHC) haplotype-related. J Neuropathol Exp Neurol. (2006) 65:1137–42. 10.1097/01.jnen.0000248547.13176.9d

Source: PubMed

3
Abonnieren