Inflammasomes are cytosolic sensors that detect pathogens and danger signals in the innate immune system. The NLRP3 inflammasome is currently the most fully characterized inflammasome and is known to detect a wide array of microbes and endogenous damage-associated molecules. Possible involvement of the NLRP3 inflammasome (or inflammasomes) in the development of multiple sclerosis (MS) was suggested in a number of studies. Recent studies showed that the NLRP3 inflammasome exacerbates experimental autoimmune encephalomyelitis (EAE), an animal model of MS, although EAE can also develop without the NLRP3 inflammasome. In this paper, we discuss the NLRP3 inflammasome in MS and EAE development. 1. Inflammasomes Inflammasomes are cytosolic sensors that detect pathogens and stresses in order to mature and secrete proinflammatory cytokines, such as interleukin-1β (IL-1β) and IL-18. Inflammasomes are expressed in phagocytes, such as macrophages and dendritic cells (DCs), and form a multiprotein complex that activates caspase-1. Assembly of inflammasomes that have clear physiological functions in vivo has been reported with relatively few NOD-like receptor (NLR) family members, such as NLRP1, NLRP3 (also called cryopyrin, CIAS1, NALP3), NLRC4 (IRAF), and AIM2 [1]. The NLRP3 inflammasome is currently the most fully characterized inflammasome and is comprised of three different proteins: NLRP3, adapter protein apoptosis-associated speck-like protein (ASC), and procaspase-1. NLRP3 protein is autorepressed by an internal interaction between the NACHT domain and leucine-rich repeats (LRRs) (Figure 1(a)) [2, 3]. Derepression of NLRP3 is essential for the interaction between NLRP3 and ASC through their Pyrin domains (PYD), followed by further interaction between ASC and procaspase-1 through CARD domains (caspase activation and recruitment domains) (Figure 1(b)). Oligomerization of the NLRP3 inflammasome heterotrimer unit leads to procaspase-1 self-cleavage to generate activated caspase-1, which processes maturation of IL-1β and IL-18 and elicits rapid release of those inflammatory cytokines by cell death termed “pyroptosis” (Figure 1(c)). Molecular mechanism by which caspase-1 mediates pyroptosis is still elusive, but is distinguished from apoptosis and necrosis [4, 5]. A molecule termed CARDINAL is known to be involved in the human NLRP3 inflammasome [6] (Figure 1(b)); but its function is unknown and there is no mouse homolog of human CARDINAL. Critical role of CARDINAL in eliciting functions of inflammasomes is questioned, because mouse inflammasomes share basic
References
[1]
K. Schroder and J. Tschopp, “The inflammasomes,” Cell, vol. 140, no. 6, pp. 821–832, 2010.
[2]
I. E. Brodsky and D. Monack, “NLR-mediated control of inflammasome assembly in the host response against bacterial pathogens,” Seminars in Immunology, vol. 21, no. 4, pp. 199–207, 2009.
[3]
J. Tschopp and K. Schroder, “NLRP3 inflammasome activation: the convergence of multiple signalling pathways on ROS production?” Nature Reviews Immunology, vol. 10, no. 3, pp. 210–215, 2010.
[4]
B. T. Cookson and M. A. Brennan, “Pro-inflammatory programmed cell death,” Trends in Microbiology, vol. 9, no. 3, pp. 113–114, 2001.
[5]
S. L. Fink and B. T. Cookson, “Apoptosis, pyroptosis, and necrosis: mechanistic description of dead and dying eukaryotic cells,” Infection and Immunity, vol. 73, no. 4, pp. 1907–1916, 2005.
[6]
L. Agostini, F. Martinon, K. Burns, M. F. McDermott, P. N. Hawkins, and J. Tschopp, “NALP3 forms an IL-1β-processing inflammasome with increased activity in Muckle-Wells autoinflammatory disorder,” Immunity, vol. 20, no. 3, pp. 319–325, 2004.
[7]
I. C. Allen, M. A. Scull, C. B. Moore et al., “The NLRP3 inflammasome mediates in vivo innate immunity to influenza a virus through recognition of viral RNA,” Immunity, vol. 30, no. 4, pp. 556–565, 2009.
[8]
S. Mariathasan, D. S. Weiss, K. Newton et al., “Cryopyrin activates the inflammasome in response to toxins and ATP,” Nature, vol. 440, no. 7081, pp. 228–232, 2006.
[9]
O. Gross, H. Poeck, M. Bscheider et al., “Syk kinase signalling couples to the Nlrp3 inflammasome for anti-fungal host defence,” Nature, vol. 459, no. 7245, pp. 433–436, 2009.
[10]
F. Martinon, V. Pétrilli, A. Mayor, A. Tardivel, and J. Tschopp, “Gout-associated uric acid crystals activate the NALP3 inflammasome,” Nature, vol. 440, no. 7081, pp. 237–241, 2006.
[11]
A. Halle, V. Hornung, G. C. Petzold et al., “The NALP3 inflammasome is involved in the innate immune response to amyloid-β,” Nature Immunology, vol. 9, no. 8, pp. 857–865, 2008.
[12]
C. Dostert, V. Pétrilli, R. van Bruggen, C. Steele, B. T. Mossman, and J. Tschopp, “Innate immune activation through Nalp3 inflammasome sensing of asbestos and silica,” Science, vol. 320, no. 5876, pp. 674–677, 2008.
[13]
V. Hornung, F. Bauernfeind, A. Halle et al., “Silica crystals and aluminum salts activate the NALP3 inflammasome through phagosomal destabilization,” Nature Immunology, vol. 9, no. 8, pp. 847–856, 2008.
[14]
D. Perregaux and C. A. Gabel, “Interleukin-1β maturation and release in response to ATP and nigericin. Evidence that potassium depletion mediated by these agents is a necessary and common feature of their activity,” Journal of Biological Chemistry, vol. 269, no. 21, pp. 15195–15203, 1994.
[15]
V. Pétrilli, S. Papin, C. Dostert, A. Mayor, F. Martinon, and J. Tschopp, “Activation of the NALP3 inflammasome is triggered by low intracellular potassium concentration,” Cell Death and Differentiation, vol. 14, no. 9, pp. 1583–1589, 2007.
[16]
T. D. Kanneganti, M. Lamkanfi, Y. G. Kim et al., “Pannexin-1-mediated recognition of bacterial molecules activates the cryopyrin inflammasome independent of toll-like receptor signaling,” Immunity, vol. 26, no. 4, pp. 433–443, 2007.
[17]
R. Zhou, A. S. Yazdi, P. Menu, and J. Tschopp, “A role for mitochondria in NLRP3 inflammasome activation,” Nature, vol. 469, pp. 221–225, 2011.
[18]
K. Nakahira, J. A. Haspel, V. A. K. Rathinam et al., “Autophagy proteins regulate innate immune responses by inhibiting the release of mitochondrial DNA mediated by the NALP3 inflammasome,” Nature Immunology, vol. 12, no. 3, pp. 222–230, 2011.
[19]
F. Martinon, “Signaling by ROS drives inflammasome activation,” European Journal of Immunology, vol. 40, no. 3, pp. 616–619, 2010.
[20]
H. M. Hoffman, J. L. Mueller, D. H. Broide, A. A. Wanderer, and R. D. Kolodner, “Mutation of a new gene encoding a putative pyrin-like protein causes familial cold autoinflammatory syndrome and Muckle-Wells syndrome,” Nature Genetics, vol. 29, no. 3, pp. 301–305, 2001.
[21]
J. Feldmann, A. M. Prieur, P. Quartier et al., “Chronic infantile neurological cutaneous and articular syndrome is caused by mutations in CIAS1, a gene highly expressed in polymorphonuclear cells and chondrocytes,” American Journal of Human Genetics, vol. 71, no. 1, pp. 198–203, 2002.
[22]
J. P. Y. Ting, D. L. Kastner, and H. M. Hoffman, “CATERPILLERs, pyrin and hereditary immunological disorders,” Nature Reviews Immunology, vol. 6, no. 3, pp. 183–195, 2006.
[23]
T. Strowig, J. Henao-Mejia, E. Elinav, and R. Flavell, “Inflammasomes in health and disease,” Nature, vol. 481, pp. 278–286, 2012.
[24]
C. Conforti-Andreoni, P. Ricciardi-Castagnoli, and A. Mortellaro, “The inflammasomes in health and disease: from genetics to molecular mechanisms of autoinflammation and beyond,” Cellular and Molecular Immunology, vol. 8, no. 2, pp. 135–145, 2011.
[25]
D. de Nardo and E. Latz, “NLRP3 inflammasomes link inflammation and metabolic disease,” Trends in Immunology, vol. 32, no. 8, pp. 373–379, 2011.
[26]
P. Menu and J. E. Vince, “The NLRP3 inflammasome in health and disease: the good, the bad and the ugly,” Clinical and Experimental Immunology, vol. 166, no. 1, pp. 1–15, 2011.
[27]
S. Compeyrot-Lacassagne, T. A. Tran, S. Guillaume-Czitrom, I. Marie, and I. Koné-Paut, “Brain multiple sclerosis-like lesions in a patient with Muckle-Wells syndrome,” Rheumatology, vol. 48, no. 12, pp. 1618–1619, 2009.
[28]
C. Dodé, N. le D?, L. Cuisset et al., “New mutations of CIAS1 that are responsible for Muckle-Wells syndrome and familial cold urticaria: a novel mutation underlies both syndromes,” American Journal of Human Genetics, vol. 70, no. 6, pp. 1498–1506, 2002.
[29]
R. Furlan, M. Filippi, A. Bergami et al., “Peripheral levels of caspase-1 mRNA correlate with disease activity in patients with multiple sclerosis; a preliminary study,” Journal of Neurology Neurosurgery and Psychiatry, vol. 67, no. 6, pp. 785–788, 1999.
[30]
X. Ming, W. Li, Y. Maeda et al., “Caspase-1 expression in multiple sclerosis plaques and cultured glial cells,” Journal of the Neurological Sciences, vol. 197, no. 1-2, pp. 9–18, 2002.
[31]
W. X. Huang, P. Huang, and J. Hillert, “Increased expression of capase-1 and interleukin-18 in peripheral blood mononuclear cells in patients with multiple sclerosis,” Multiple Sclerosis, vol. 10, no. 5, pp. 482–487, 2004.
[32]
NCBI-dbSNP Website.
[33]
B. A. de Jong, T. W. J. Huizinga, E. L. E. M. Bollen et al., “Production of IL-1β and IL-1Ra as risk factors for susceptibility and progression of relapse-onset multiple sclerosis,” Journal of Neuroimmunology, vol. 126, no. 1-2, pp. 172–179, 2002.
[34]
J. B. A. Crusius, A. S. Pena, B. W. van Oosten et al., “Interleukin-1 receptor antagonist gene polymorphism and multiple sclerosis,” Lancet, vol. 346, no. 8980, pp. 979–980, 1995.
[35]
R. Voltz, M. Hartmann, S. Spuler et al., “Multiple sclerosis: longitudinal measurement of interleukin-1 receptor antagonist,” Journal of Neurology Neurosurgery and Psychiatry, vol. 62, no. 2, pp. 200–201, 1997.
[36]
E. G. de la Concha, R. Arroyo, J. B. A. Crusius et al., “Combined effect of HLA-DRB1*1501 and interleukin-1 receptor antagonist gene allele 2 in susceptibility to relapsing/remitting multiple sclerosis,” Journal of Neuroimmunology, vol. 80, no. 1-2, pp. 172–178, 1997.
[37]
R. Feakes, S. Sawcer, S. Broadley et al., “Interleukin 1 receptor antagonist (IL-1ra) in multiple sclerosis,” Journal of Neuroimmunology, vol. 105, no. 1, pp. 96–101, 2000.
[38]
O. H. Kantarci, E. J. Atkinson, D. D. Hebrink, C. T. McMurray, and B. G. Weinshenker, “Association of two variants in IL-1β and IL-1 receptor antagonist genes with multiple sclerosis,” Journal of Neuroimmunology, vol. 106, no. 1-2, pp. 220–227, 2000.
[39]
M. Luomala, T. Lehtim?ki, I. Elovaara et al., “A study of interleukin-1 cluster genes in susceptibility to and severity of multiple sclerosis,” Journal of the Neurological Sciences, vol. 185, no. 2, pp. 123–127, 2001.
[40]
M. Niino, S. Kikuchi, T. Fukazawa, I. Yabe, H. Sasaki, and K. Tashiro, “Genetic polymorphisms of IL-1β and IL-1 receptor antagonist in association with multiple sclerosis in Japanese patients,” Journal of Neuroimmunology, vol. 118, no. 2, pp. 295–299, 2001.
[41]
D. Burger, N. Molnarfi, M. S. Weber et al., “Glatiramer acetate increases IL-1 receptor antagonist but decreases T cell-induced IL-1β in human monocytes and multiple sclerosis,” Proceedings of the National Academy of Sciences of the United States of America, vol. 106, no. 11, pp. 4355–4359, 2009.
[42]
F. Nicoletti, F. Patti, R. di Marco et al., “Circulating serum levels of IL-1ra in patients with relapsing remitting multiple sclerosis are normal during remission phases but significantly increased either during exacerbations or in response to IFN-β treatment,” Cytokine, vol. 8, no. 5, pp. 395–400, 1996.
[43]
Y. Yiangou, P. Facer, P. Durrenberger et al., “COX-2, CB2 and P2X7-immunoreactivities are increased in activated microglial cells/macrophages of multiple sclerosis and amyotrophic lateral sclerosis spinal cord,” BMC Neurology, vol. 6, article 12, 2006.
[44]
C. Matute, “Interaction between glutamate signalling and immune attack in damaging oligodendrocytes,” Neuron Glia Biology, vol. 3, no. 4, pp. 281–285, 2007.
[45]
M. Caragnano, P. Tortorella, A. Bergami, et al., “Monocytes P2X7 purinergic receptor is modulated by glatiramer acetate in multiple sclerosis,” Journal of Neuroimmunology, vol. 245, pp. 93–97, 2012.
[46]
O. Oyanguren-Desez, A. Rodriguez-Antiguedad, P. Villoslada, M. Domercq, E. Alberdi, and C. Matute, “Gain-of-function of P2X7 receptor gene variants in multiple sclerosis,” Cell Calcium, vol. 50, pp. 468–472, 2011.
[47]
A. M. Amorini, A. Petzold, B. Tavazzi et al., “Increase of uric acid and purine compounds in biological fluids of multiple sclerosis patients,” Clinical Biochemistry, vol. 42, no. 10-11, pp. 1001–1006, 2009.
[48]
B. Liu, Y. Shen, K. Xiao, Y. Tang, L. Cen, and J. Wei, “Serum uric acid levels in patients with multiple sclerosis: a meta-analysis,” Neurological Research, vol. 34, no. 2, pp. 163–171, 2012.
[49]
C. T. Bever Jr. and D. W. Garver, “Increased cathepsin B activity in multiple sclerosis brain,” Journal of the Neurological Sciences, vol. 131, no. 1, pp. 71–73, 1995.
[50]
M. Inoue, K. L. Williams, T. Oliver et al., “Interferon-β therapy against EAE is effective only when development of the disease depends on the NLRP3 inflammasome,” Science Signaling, vol. 5, no. 225, p. ra38, 2012.
[51]
D. Gris, Z. Ye, H. A. Iocca et al., “NLRP3 plays a critical role in the development of experimental autoimmune encephalomyelitis by mediating Th1 and Th17 responses,” Journal of Immunology, vol. 185, no. 2, pp. 974–981, 2010.
[52]
M. Inoue, K. L. Williams, M. D. Gunn, and M. L. Shinohara, “NLRP3 inflammasome induces chemotactic immune cell migration to the CNS in experimental autoimmune encephalomyelitis,” Proceedings of the National Academy of Sciences of the United States of America, vol. 109, no. 26, pp. 10480–10485, 2012.
[53]
S. Jha, S. Y. Srivastava, W. J. Brickey et al., “The inflammasome sensor, NLRP3, regulates CNS inflammation and demyelination via caspase-1 and interleukin-18,” Journal of Neuroscience, vol. 30, no. 47, pp. 15811–15820, 2010.
[54]
R. Furlan, G. Martino, F. Galbiati et al., “Caspase-1 regulates the inflammatory process leading to autoimmune demyelination,” Journal of Immunology, vol. 163, no. 5, pp. 2403–2409, 1999.
[55]
Z. Ahmed, A. I. Doward, G. Pryce et al., “A role for caspase-1 and -3 in the pathology of experimental allergic encephalomyelitis: inflammation versus degeneration,” American Journal of Pathology, vol. 161, no. 5, pp. 1577–1586, 2002.
[56]
C. Sutton, C. Brereton, B. Keogh, K. H. G. Mills, and E. C. Lavelle, “A crucial role for interleukin (IL)-1 in the induction of IL-17-producing T cells that mediate autoimmune encephalomyelitis,” Journal of Experimental Medicine, vol. 203, no. 7, pp. 1685–1691, 2006.
[57]
V. Badovinac, M. Mostarica-Stojkovi?, C. A. Dinarello, and S. Sto?i?-Gruji?i?, “Interleukin-1 receptor antagonist suppresses experimental autoimmune encephalomyelitis (EAE) in rats by influencing the activation and proliferation of encephalitogenic cells,” Journal of Neuroimmunology, vol. 85, no. 1, pp. 87–95, 1998.
[58]
F. D. Shi, K. Takeda, S. Akira, N. Sarvetnick, and H. G. Ljunggren, “IL-18 directs autoreactive T cells and promotes autodestruction in the central nervous system via induction of IFN-γ by NK cells,” Journal of Immunology, vol. 165, no. 6, pp. 3099–3104, 2000.
[59]
C. Matute, I. Torre, F. Pérez-Cerdá et al., “P2X7 receptor blockade prevents ATP excitotoxicity in oligodendrocytes and ameliorates experimental autoimmune encephalomyelitis,” Journal of Neuroscience, vol. 27, no. 35, pp. 9525–9533, 2007.
[60]
J. I. Alvarez, A. Dodelet-Devillers, H. Kebir et al., “The hedgehog pathway promotes blood-brain barrier integrity and CNS immune quiescence,” Science, vol. 334, no. 6063, pp. 1727–1731, 2011.
[61]
C. C. Ferrari, A. M. Depino, F. Prada et al., “Reversible demyelination, blood-brain barrier breakdown, and pronounced neutrophil recruitment induced by chronic IL-1 expression in the brain,” American Journal of Pathology, vol. 165, no. 5, pp. 1827–1837, 2004.
[62]
C. E. Sutton, S. J. Lalor, C. M. Sweeney, C. F. Brereton, E. C. Lavelle, and K. H. G. Mills, “Interleukin-1 and IL-23 Induce Innate IL-17 Production from γδ T Cells, Amplifying Th17 Responses and Autoimmunity,” Immunity, vol. 31, no. 2, pp. 331–341, 2009.
[63]
S. Jander and G. Stoll, “Differential induction of interleukin-12, interleukin- 18, and interleukin-1β converting enzyme mRNA in experimental autoimmune encephalomyelitis of the Lewis rat,” Journal of Neuroimmunology, vol. 91, no. 1-2, pp. 93–99, 1998.
[64]
G. Wildbaum, S. Youssef, N. Grabie, and N. Karin, “Neutralizing antibodies to IFN-γ-inducing factor prevent experimental autoimmune encephalomyelitis,” Journal of Immunology, vol. 161, no. 11, pp. 6368–6374, 1998.
[65]
I. Gutcher, E. Urich, K. Wolter, M. Prinz, and B. Becher, “Interleukin 18-independent engagement of interleukin 18 receptor-α is required for autoimmune inflammation,” Nature Immunology, vol. 7, no. 9, pp. 946–953, 2006.
[66]
S. J. Lalor, L. S. Dungan, C. E. Sutton, S. A. Basdeo, J. M. Fletcher, and K. H. G. Mills, “Caspase-1-processed cytokines IL-1β and IL-18 promote IL-17 production by γδ and CD4 T cells that mediate autoimmunity,” Journal of Immunology, vol. 186, no. 10, pp. 5738–5748, 2011.
[67]
B. T. Fife, G. B. Huffnagle, W. A. Kuziel, and W. J. Karpus, “CC chemokine receptor 2 is critical for induction of experimental autoimmune encephalomyelitis,” Journal of Experimental Medicine, vol. 192, no. 6, pp. 899–905, 2000.
[68]
D. R. Huang, J. Wang, P. Kivisakk, B. J. Rollins, and R. M. Ransohoff, “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,” Journal of Experimental Medicine, vol. 193, no. 6, pp. 713–725, 2001.
[69]
T. L. S?rensen and F. Sellebjerg, “Distinct chemokine receptor and cytokine expression profile in secondary progressive MS,” Neurology, vol. 57, no. 8, pp. 1371–1376, 2001.
[70]
T. Misu, H. Onodera, K. Fujihara et al., “Chemokine receptor expression on T cells in blood and cerebrospinal fluid at relapse and remission of multiple sclerosis: imbalance of Th1/Th2-associated chemokine signaling,” Journal of Neuroimmunology, vol. 114, no. 1-2, pp. 207–212, 2001.
[71]
C. McManus, J. W. Berman, F. M. Brett, H. Staunton, M. Farrell, and C. F. Brosnan, “MCP-1, MCP-2 and MCP-3 expression in multiple sclerosis lesions: an immunohistochemical and in situ hybridization study,” Journal of Neuroimmunology, vol. 86, no. 1, pp. 20–29, 1998.
[72]
I. Nelissen, B. Dubois, A. Goris, I. Ronsse, H. Carton, and G. Opdenakker, “Gelatinase B, PECAM-1 and MCP-3 gene polymorphisms in Belgian multiple sclerosis,” Journal of the Neurological Sciences, vol. 200, no. 1-2, pp. 43–48, 2002.
[73]
J. V. Kim, N. Jiang, C. E. Tadokoro et al., “Two-photon laser scanning microscopy imaging of intact spinal cord and cerebral cortex reveals requirement for CXCR6 and neuroinflammation in immune cell infiltration of cortical injury sites,” Journal of Immunological Methods, vol. 352, no. 1-2, pp. 89–100, 2010.
[74]
N. Fukumoto, T. Shimaoka, H. Fujimura et al., “Critical roles of CXC chemokine ligand 16/scavenger receptor that binds phosphatidylserine and oxidized lipoprotein in the pathogenesis of both acute and adoptive transfer experimental autoimmune encephalomyelitis,” Journal of Immunology, vol. 173, no. 3, pp. 1620–1627, 2004.
[75]
T. Uede, “Osteopontin, intrinsic tissue regulator of intractable inflammatory diseases,” Pathology International, vol. 61, no. 5, pp. 265–280, 2011.
[76]
D. Chabas, S. E. Baranzini, D. Mitchell et al., “The influence of the proinflammatory cytokine, osteopontin, on autoimmue demyelinating desease,” Science, vol. 294, no. 5547, pp. 1731–1735, 2001.
[77]
M. Jansson, V. Panoutsakopoulou, J. Baker, L. Klein, and H. Cantor, “Cutting edge: attenuated experimental autoimmune encephalomyelitis in ETA-1/osteopontin-deficient mice,” Journal of Immunology, vol. 168, no. 5, pp. 2096–2099, 2002.
[78]
M. L. Shinohara, J. H. Kim, V. A. Garcia, and H. Cantor, “Engagement of the type I interferon receptor on dendritic cells inhibits T helper 17 cell development: role of intracellular osteopontin,” Immunity, vol. 29, no. 1, pp. 68–78, 2008.
[79]
E. M. Hur, S. Youssef, M. E. Haws, S. Y. Zhang, R. A. Sobel, and L. Steinman, “Osteopontin-induced relapse and progression of autoimmune brain disease through enhanced survival of activated T cells,” Nature Immunology, vol. 8, no. 1, pp. 74–83, 2007.
[80]
M. L. Shinohara, M. Jansson, E. S. Hwang, M. B. F. Werneck, L. H. Glimcher, and H. Cantor, “T-bet-dependent expression of osteopontin contributes to T cell polarization,” Proceedings of the National Academy of Sciences of the United States of America, vol. 102, no. 47, pp. 17101–17106, 2005.
[81]
G. Murugaiyan, A. Mittal, and H. L. Weiner, “Increased osteopontin expression in dendritic cells amplifies IL-17 production by CD4+ T cells in experimental autoimmune encephalomyelitis and in multiple sclerosis,” Journal of immunology, vol. 181, no. 11, pp. 7480–7488, 2008.
[82]
H. Cantor and M. L. Shinohara, “Regulation of T-helper-cell lineage development by osteopontin: the inside story,” Nature Reviews Immunology, vol. 9, no. 2, pp. 137–141, 2009.
[83]
T. A. Yednock, C. Cannon, L. C. Fritz, F. Sanchez-Madrid, L. Steinmann, and N. Karin, “Prevention of experimental autoimmune encephalomyelitis by antibodies against α4β1 integrin,” Nature, vol. 356, no. 6364, pp. 63–66, 1992.
[84]
L. Steinman, “Blocking adhesion molecules as therapy for multiple sclerosis: natalizumab,” Nature Reviews Drug Discovery, vol. 4, no. 6, pp. 510–518, 2005.
[85]
P. J. Shaw, J. R. Lukens, S. Burns, H. Chi, M. A. McGargill, and T. D. Kanneganti, “Cutting edge: critical role for PYCARD/ASC in the development of experimental autoimmune encephalomyelitis,” Journal of Immunology, vol. 184, no. 9, pp. 4610–4614, 2010.