Role of Innate Immunity in Multiple Sclerosis ws

Editorial

Austin J Mult Scler & Neuroimmunol. 2015;2(1): 1007.

Role of Innate Immunity in Multiple Sclerosis

Koudriavtseva T1* and Mainero C2

1Multiple Sclerosis Center, Neurology Unit, Regina Elena National Cancer Institute, Italy

2Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, USA

*Corresponding author: Tatiana Koudriavtseva, Multiple Sclerosis Center, Neurology Unit, Regina Elena National Cancer Institute, IFO, Via Elio Chianesi 53, 00144 - Rome, Italy

Received: December 29, 2014; Accepted: January 28, 2015; Published: January 30, 2015

Editorial

Multiple sclerosis (MS) is a chronic demyelinating inflammatory disease of the central nervous system (CNS) affecting prevalently young adults and with multi-factorial pathogenesis that includes genetic and environmental factors [1-3]. The pathogenesis of MS has been long attributed to self-reactive T cells but recently the relevant role of B cells, another component of adaptive immunity, has been recognized [4]. It has been demonstrated that innate immunity also contributes significantly to MS pathogenesis both in the initial and in the advanced stages of the disease [5-8]. In particular, activation of microglial cells, which physiologically act as cleaners of the brain microenvironment in response to injury and infections, has been widely reported in both white matter and gray matter tissue in MS [3]. Microglial activation in the absence of lymphocytes or myelin phagocytosis, has been observed in early MS lesions [9,10], expression of a primary involvement of innate immunity. Likewise, predominant microglia activation into newly formed cortical lesions has been detected in the progressive MS phase [11]. Widespread damage of normal appearing white matter (NAWM) seems to be related to chronic microglial activation with marked expression of i-NOS and myeloperoxidase [3]. Moreover, it has been hypothesized that the progressive phase of MS could be mainly driven by the innate immune system contributing to the neurodegenerative changes of the disease [7]. In vivo positron emission tomography studies using radioligands that selectively target the translocator protein (TSPO), expressed in activated microglia and macrophages, have recently demonstrated increased TSPO expression in NAWM and cortex of patients from the earliest to the progressive MS stages [12].

Innate immunity represents the immediate, nonspecific defense against infections and dangerous agents. Adaptive immunity develops a specific immunological memory after the first contact with a pathogen, exponentially enhancing its successive responses [13]. There are close interrelationships between both types of immunity, and innate immunity stimulates and modulates the adaptive one. Innate immunity acts through its essential processes such as inflammation and blood coagulation [14-17]. The coagulation cascade has had primordially the function of limiting invasiveness of potential pathogens by trapping them in the fibrin network [15]. Physiologically, coagulant processes are balanced by the natural anticoagulant system needed to limit the host damage, and the lack of homeostatic interactions between these systems leads to thrombosis [14-17]. This could occur in people with genetic or environmentinduced impaired anti-coagulant, and/or anti-oxidative system or during prolonged inflammatory-thrombotic processes. In any case, the presence of thrombotic events indicates an excessive activation of innate immunity.

Since extensive reviews have already described in detail the involvement of innate immunity cells in MS and in experimental allergic encephalomyelitis (EAE) [2,5-8], this editorial aims to emphasize also the important role of the coagulant component of innate immunity in MS and EAE. It is not negligible that such component could be an easily reachable target for a possible therapeutic intervention.

In fact, while many efforts have been aimed to better define the function of innate immune cells in MS [6,7], the role and impact of the coagulant component of innate immunity are still unclear, though there is ample literature on the presence of systemic thrombosis in MS [18-23]. Furthermore, recent studies provided evidence for involvement of platelets [24-26] and complement [27-29] in MS, which participate in the innate immune response by linking inflammation and coagulation.

In addition to systemic thrombosis, several data in the literature have shown the presence of brain thrombotic processes in MS and in EAE. In 1935, Putnam had already considered the role of venule thrombosis in CNS demyelination [30]. A pathological study in acute MS reported fibrin deposition on endothelial cells in many thin veins and capillaries, with some thrombosed vessels, in areas without myelin damage or reactive parenchyma changes [31]. The areas of microglial activation associated with fibrin deposition were found in acute or early MS and in rat EAE, representing a first stage of tissue injury before active demyelination and massive T-cell infiltration [9]. Coagulation proteins were highlighted within the chronic active plaque in MS by the proteomic analysis [32]. In vivo, some proteins involved in coagulation such as β2glicoprotein I, fibrinogen and complement C4, were found in most abundant quantities in the cerebrospinal fluid of fulminant MS compared to controls [33]. Another paper reported an increase of soluble thrombomodulin in relapsing-remitting MS, released from the surface of damaged cerebrovascular endothelial cells, reducing their normal function in promoting the activation of anticoagulant protein C [34]. This leads to a reduction in the inhibitory function of protein C on inflammatory cell migration across the blood-brain barrier (BBB).

Also in EAE, fibrin deposition preceded and regulated inflammatory demyelination, while its genetic or pharmacologic depletion ameliorated both clinical symptoms and inflammatory response [35]. It has been demonstrated that early perivascular microglial clustering, triggered by fibrinogen leakage after BBB disruption, contributed to axonal damage in EAE before myelin loss or paralysis onset, and that this process was blocked by anticoagulant treatment or by genetic deletion of fibrinogen [36]. Likewise, hirudin or recombinant activated protein C (rAPC) improved EAE and suppressed pro-inflammatory T-helper1 and T-helper17 cytokines in astrocytes and immune cells [32]. Both anticoagulant and signaling functions of rAPC have been demonstrated necessary for improving EAE [32].

Chapman stressed the importance of thrombin in inflammatory brain diseases [37,38]. Thrombin converts circulating fibrinogen to fibrin, and has numerous hormone-like functions affecting, among others, microglia and astrocytes [39]. The activity of thrombin in the brain is regulated by endogenous thrombin inhibitors such as serum antithrombin III, expressed in the liver and less in the brain, and brain protease nexin-1 (PN-1) secreted by glial cells and neurons [37,38]. It has been demonstrated that the plasma thrombin-antithrombin complexes were associated with EAE severity, increasing immediately prior to EAE symptoms and decreasing in relation to their improvement [40]. Similarly, an increase of brain PN-1 has been shown at the preclinical stage in mouse EAE [41]. The suppression of EAE by dermatan sulfate [42] or low doses of heparins [43] has been also demonstrated, as a result Chapman proposed thrombin as a therapeutic target in MS [37].

In conclusion, involvement of the coagulant component of innate immunity in MS is largely supported by several studies in EAE and MS. Therapeutic targeting of innate immunity, both its cellular and coagulant components, could be a promising approach for treating MS.

References

  1. Noseworthy JH, Lucchinetti C, Rodriguez M, Weinshenker BG. Multiple sclerosis. N Engl J Med. 2000; 343: 938-952.
  2. Sospedra M, Martin R. Immunology of multiple sclerosis. Annu Rev Immunol. 2005; 23: 683-747.
  3. Lassmann H, Brück W, Lucchinetti CF. The immunopathology of multiple sclerosis: an overview. Brain Pathol. 2007; 17: 210-218.
  4. Disanto G, Morahan JM, Barnett MH, Giovannoni G, Ramagopalan SV. The evidence for a role of B cells in multiple sclerosis. Neurology. 2012; 78: 823-832.
  5. Weiner HL. A shift from adaptive to innate immunity: a potential mechanism of disease progression in multiple sclerosis. J Neurol. 2008; 255 Suppl 1: 3-11.
  6. Gandhi R, Laroni A, Weiner HL. Role of the innate immune system in the pathogenesis of multiple sclerosis. J Neuroimmunol. 2010; 221: 7-14.
  7. Mayo L, Quintana FJ, Weiner HL. The innate immune system in demyelinating disease. Immunol Rev. 2012; 248: 170-187.
  8. Hernández-Pedro NY, Espinosa-Ramirez G, de la Cruz VP, Pineda B, Sotelo J. Initial immunopathogenesis of multiple sclerosis: innate immune response. Clin Dev Immunol. 2013; 2013: 413465.
  9. Marik C, Felts PA, Bauer J, Lassmann H, Smith KJ. Lesion genesis in a subset of patients with multiple sclerosis: a role for innate immunity? Brain. 2007; 130: 2800-2815.
  10. Barnett MH, Prineas JW. Relapsing and remitting multiple sclerosis: pathology of the newly forming lesion. Ann Neurol. 2004; 55: 458-468.
  11. Bø L, Vedeler CA, Nyland H, Trapp BD, Mørk SJ. Intracortical multiple sclerosis lesions are not associated with increased lymphocyte infiltration. Mult Scler. 2003; 9: 323–331.
  12. Giannì C, Govindarajan ST, Fan AP, Louapre C, Loggia M, Catana C, et al. [11C]-PBR28 MR-PET imaging detects in vivo inflammation in normal appearing white matter and cortical sulci in multiple sclerosis. ECTRIMS-ACTRIMS, Boston, USA. 2014.
  13. Abbas AK, Lichtman AH, Pillai S. Basic Immunology, functions and disorders of the immune system. 4th ed. Philadelphia: Saunders. 2012.
  14. Esmon CT. Interactions between the innate immune and blood coagulation systems. Trends Immunol. 2004; 25: 536-542.
  15. Esmon CT, Xu J, Lupu F. Innate immunity and coagulation. J Thromb Haemost. 2011; 9 Suppl 1: 182-188.
  16. Esmon CT. Protein C anticoagulant system--anti-inflammatory effects. Semin Immunopathol. 2012; 34: 127-132.
  17. Danckwardt S, Hentze MW, Kulozik AE. Pathologies at the nexus of blood coagulation and inflammation: thrombin in hemostasis, cancer, and beyond. J Mol Med (Berl). 2013; 91: 1257-1271.
  18. Christiansen CF, Christensen S, Farkas DK, Miret M, Sørensen HT, Pedersen L. Risk of arterial cardiovascular diseases in patients with multiple sclerosis: a population-based cohort study. Neuroepidemiology. 2010; 35: 267-274.
  19. Christensen S, Farkas DK, Pedersen L, Miret M, Christiansen CF, Sørensen HT. Multiple sclerosis and risk of venous thromboembolism: a population-based cohort study. Neuroepidemiology. 2012; 38: 76-83.
  20. Christiansen CF. Risk of vascular disease in patients with multiple sclerosis: a review. Neurol Res. 2012; 34: 746-753.
  21. Zöller B, Li X, Sundquist J, Sundquist K. Autoimmune diseases and venous thromboembolism: a review of the literature. Am J Cardiovasc Dis. 2012; 2: 171-183.
  22. Ramagopalan SV, Wotton CJ, Handel AE, Yeates D, Goldacre MJ. Risk of venous thromboembolism in people admitted to hospital with selected immune-mediated diseases: record-linkage study. BMC Med. 2011; 9: 1.
  23. Ocak G, Vossen CY, Verduijn M, Dekker FW, Rosendaal FR, Cannegieter SC, et al. Risk of venous thrombosis in patients with major illnesses: results from the MEGA study. J Thromb Haemost. 2013; 11: 116-123.
  24. Horstman LL, Jy W, Ahn YS, Zivadinov R, Maghzi AH, Etemadifar M, et al. Role of platelets in neuroinflammation: a wide-angle perspective. J Neuroinflammation. 2010; 7: 10.
  25. Nurden AT. Platelets, inflammation and tissue regeneration. Thromb Haemost. 2011; 105 Suppl 1: S13-33.
  26. Behari M, Shrivastava M. Role of platelets in neurodegenerative diseases: a universal pathophysiology. Int J Neurosci. 2013; 123: 287-299.
  27. Horstman LL, Jy W, Ahn YS, Maghzi AH, Etemadifar M, Alexander JS, et al. Complement in neurobiology. Front Biosci (Landmark Ed). 2011; 16: 2921-2960.
  28. Veerhuis R, Nielsen HM, Tenner AJ. Complement in the brain. Mol Immunol. 2011; 48: 1592-1603.
  29. Ingram G, Hakobyan S, Hirst CL, Harris CL, Loveless S, Mitchell JP, et al. Systemic complement profiling in multiple sclerosis as a biomarker of disease state. Mult Scler. 2012; 18: 1401-1411.
  30. Putnam TJ. Studies in multiple sclerosis: encephalitis and sclerotic plaques produced by venular obstruction. Archives of Neurology and Psychiatry 1935. 33: 929-940.
  31. Wakefield AJ, More LJ, Difford J, McLaughlin JE. Immunohistochemical study of vascular injury in acute multiple sclerosis. J Clin Pathol. 1994; 47: 129-133.
  32. Han MH, Hwang SI, Roy DB, Lundgren DH, Price JV, Ousman SS, et al. Proteomic analysis of active multiple sclerosis lesions reveals therapeutic targets. Nature. 2008; 451: 1076-1081.
  33. Füvesi J, Hanrieder J, Bencsik K, Rajda C, Kovács SK, Kaizer L, et al. Proteomic analysis of cerebrospinal fluid in a fulminant case of multiple sclerosis. Int J Mol Sci. 2012; 13: 7676-7693.
  34. Festoff BW, Li C, Woodhams B, Lynch S. Soluble thrombomodulin levels in plasma of multiple sclerosis patients and their implication. J Neurol Sci. 2012; 323: 61-65.
  35. Akassoglou K, Adams RA, Bauer J, Mercado P, Tseveleki V, Lassmann H, et al. Fibrin depletion decreases inflammation and delays the onset of demyelination in a tumor necrosis factor transgenic mouse model for multiple sclerosis. Proc Natl Acad Sci U S A. 2004; 101: 6698-6703.
  36. Davalos D, Ryu JK, Merlini M, Baeten KM, Le Moan N, Petersen MA, et al. Fibrinogen-induced perivascular microglial clustering is required for the development of axonal damage in neuroinflammation. Nat Commun. 2012; 3: 1227.
  37. Chapman J. Thrombin in inflammatory brain diseases. Autoimmun Rev. 2006; 5: 528-531.
  38. Chapman J. Coagulation in inflammatory diseases of the central nervous system. Semin Thromb Hemost. 2013; 39: 876-880.
  39. Coughlin SR. Thrombin signalling and protease-activated receptors. Nature. 2000; 407: 258-264.
  40. Inaba Y, Ichikawa M, Inoue A, Itoh M, Kyogashima M, Sekiguchi Y, et al. Plasma thrombin-antithrombin III complex is associated with the severity of experimental autoimmune encephalomyelitis. J Neurol Sci. 2001; 185: 89-93.
  41. Beilin O, Karussis DM, Korczyn AD, Gurwitz D, Aronovich R, Hantai D, et al. Increased thrombin inhibition in experimental autoimmune encephalomyelitis. J Neurosci Res. 2005; 79: 351-359.
  42. Inaba Y, Ichikawa M, Koh CS, Inoue A, Itoh M, Kyogashima M, et al. Suppression of experimental autoimmune encephalomyelitis by dermatan sulfate. Cell Immunol. 1999; 198: 96-102.
  43. Lider O1, Baharav E, Mekori YA, Miller T, Naparstek Y, Vlodavsky I, et al. Suppression of experimental autoimmune diseases and prolongation of allograft survival by treatment of animals with low doses of heparins. J Clin Invest. 1989; 83: 752-756.

Download PDF

Citation: Koudriavtseva T and Mainero C. Role of Innate Immunity in Multiple Sclerosis. Austin J Mult Scler & Neuroimmunol. 2015;2(1): 1007.

Home
Journal Scope
Online First
Current Issue
Editorial Board
Instruction for Authors
Submit Your Article
Contact Us