Sex-specific control of central nervous system autoimmunity by p38 mitogen-activated protein kinase signaling in myeloid cells
Dimitry N. Krementsov PhD
Department of Medicine, Immunobiology Program, University of Vermont, Burlington, VT
Search for more papers by this authorRajkumar Noubade PhD
Department of Medicine, Immunobiology Program, University of Vermont, Burlington, VT
Genentech, Inc, South San Francisco, CA
Search for more papers by this authorJulie A. Dragon PhD
Department of Microbiology and Molecular Genetics, University of Vermont, Burlington, VT
Search for more papers by this authorKinya Otsu MD, PhD
Cardiovascular Division, King's College London, London, UK
Search for more papers by this authorMercedes Rincon PhD
Department of Medicine, Immunobiology Program, University of Vermont, Burlington, VT
Search for more papers by this authorCorresponding Author
Cory Teuscher PhD
Department of Medicine, Immunobiology Program, University of Vermont, Burlington, VT
Address correspondence to Dr Teuscher, Immunobiology Program, C331 Given Medical Building, University of Vermont, Burlington, VT 05405. E-mail: [email protected]Search for more papers by this authorDimitry N. Krementsov PhD
Department of Medicine, Immunobiology Program, University of Vermont, Burlington, VT
Search for more papers by this authorRajkumar Noubade PhD
Department of Medicine, Immunobiology Program, University of Vermont, Burlington, VT
Genentech, Inc, South San Francisco, CA
Search for more papers by this authorJulie A. Dragon PhD
Department of Microbiology and Molecular Genetics, University of Vermont, Burlington, VT
Search for more papers by this authorKinya Otsu MD, PhD
Cardiovascular Division, King's College London, London, UK
Search for more papers by this authorMercedes Rincon PhD
Department of Medicine, Immunobiology Program, University of Vermont, Burlington, VT
Search for more papers by this authorCorresponding Author
Cory Teuscher PhD
Department of Medicine, Immunobiology Program, University of Vermont, Burlington, VT
Address correspondence to Dr Teuscher, Immunobiology Program, C331 Given Medical Building, University of Vermont, Burlington, VT 05405. E-mail: [email protected]Search for more papers by this authorAbstract
Objective
Multiple sclerosis (MS) is a chronic inflammatory demyelinating disease of the central nervous system (CNS), characterized by a global increasing incidence driven by relapsing–remitting disease in females. Investigators have described p38 mitogen-activated protein kinase (MAPK) as a key regulator of inflammatory responses in autoimmunity, but its role in the sexual dimorphism in MS or MS models remains unexplored.
Methods
Toward this end, we used experimental autoimmune encephalomyelitis (EAE), the principal animal model of MS, combined with pharmacologic and genetic inhibition of p38 MAPK activity and transcriptomic analyses.
Results
Pharmacologic inhibition of p38 MAPK selectively ameliorated EAE in female mice. Conditional deletion studies demonstrated that p38α signaling in macrophages/myeloid cells, but not T cells or dendritic cells, mediated this sexual dimorphism, which was dependent on the presence of adult sex hormones. Analysis of CNS inflammatory infiltrates showed that female but not male mice lacking p38α in myeloid cells exhibited reduced immune cell activation compared with controls, whereas peripheral T-cell priming was unaffected in both sexes. Transcriptomic analyses of myeloid cells revealed differences in p38α-controlled transcripts comprising female- and male-specific gene modules, with greater p38α dependence of proinflammatory gene expression in females.
Interpretation
Our findings demonstrate a key role for p38α in myeloid cells in CNS autoimmunity and uncover important molecular mechanisms underlying sex differences in disease pathogenesis. Taken together, our results suggest that the p38 MAPK signaling pathway represents a novel target for much needed disease-modifying therapies for MS. Ann Neurol 2014;75:50–66
Supporting Information
Additional Supporting Information may be found in the online version of this article.
Filename | Description |
---|---|
ana24020-sup-0001-suppinfo.docx113.4 KB | Supplementary Information |
Please note: The publisher is not responsible for the content or functionality of any supporting information supplied by the authors. Any queries (other than missing content) should be directed to the corresponding author for the article.
References
- 1 Ramagopalan SV, Sadovnick AD. Epidemiology of multiple sclerosis. Neurol Clin 2011; 29: 207–217.
- 2 Greenstein JI. Current concepts of the cellular and molecular pathophysiology of multiple sclerosis. Dev Neurobiol 2007; 67: 1248–1265.
- 3 Sawcer S, Hellenthal G, Pirinen M, et al. Genetic risk and a primary role for cell-mediated immune mechanisms in multiple sclerosis. Nature 2011; 476: 214–219.
- 4 Ebers GC. Environmental factors and multiple sclerosis. Lancet Neurol 2008; 7: 268–277.
- 5 Rincon M, Davis RJ. Regulation of the immune response by stress-activated protein kinases. Immunol Rev 2009; 228: 212–224.
- 6 Lee JC, Laydon JT, McDonnell PC, et al. A protein kinase involved in the regulation of inflammatory cytokine biosynthesis. Nature 1994; 372: 739–746.
- 7 Liverton NJ, Butcher JW, Claiborne CF, et al. Design and synthesis of potent, selective, and orally bioavailable tetrasubstituted imidazole inhibitors of p38 mitogen-activated protein kinase. J Med Chem 1999; 42: 2180–2190.
- 8 Hollenbach E, Neumann M, Vieth M, et al. Inhibition of p38 MAP kinase- and RICK/NF-kappaB-signaling suppresses inflammatory bowel disease. FASEB J 2004; 18: 1550–1552.
- 9 Ando H, Kurita S, Takamura T. The specific p38 mitogen-activated protein kinase pathway inhibitor FR167653 keeps insulitis benign in nonobese diabetic mice. Life Sci 2004; 74: 1817–1827.
- 10 Genovese MC. Inhibition of p38: has the fat lady sung? Arthritis Rheum 2009; 60: 317–320.
- 11 Hammaker D, Firestein GS. “Go upstream, young man”: lessons learned from the p38 saga. Ann Rheum Dis 2010; 69(suppl 1): i77–i82.
- 12 Cho JH, Gregersen PK. Genomics and the multifactorial nature of human autoimmune disease. N Engl J Med 2011; 365: 1612–1623.
- 13 Shin T, Ahn M, Jung K, et al. Activation of mitogen-activated protein kinases in experimental autoimmune encephalomyelitis. J Neuroimmunol 2003; 140: 118–125.
- 14 Lock C, Hermans G, Pedotti R, et al. Gene-microarray analysis of multiple sclerosis lesions yields new targets validated in autoimmune encephalomyelitis. Nat Med 2002; 8: 500–508.
- 15 Lu L, Wang J, Zhang F, et al. Role of SMAD and non-SMAD signals in the development of Th17 and regulatory T cells. J Immunol 2010; 184: 4295–4306.
- 16 Noubade R, Krementsov DN, Del Rio R, et al. Activation of p38 MAPK in CD4 T cells controls IL-17 production and autoimmune encephalomyelitis. Blood 2011; 118: 3290–3300.
- 17 Namiki K, Matsunaga H, Yoshioka K, et al. Mechanism for p38alpha-mediated experimental autoimmune encephalomyelitis. J Biol Chem 2012; 287: 24228–24238.
- 18 Huang G, Wang Y, Vogel P, et al. Signaling via the kinase p38alpha programs dendritic cells to drive TH17 differentiation and autoimmune inflammation. Nat Immunol 2012; 13: 152–161.
- 19 Jirmanova L, Giardino Torchia ML, Sarma ND, et al. Lack of the T cell-specific alternative p38 activation pathway reduces autoimmunity and inflammation. Blood 2011; 118: 3280–3289.
- 20 Guo X, Harada C, Namekata K, et al. Regulation of the severity of neuroinflammation and demyelination by TLR-ASK1-p38 pathway. EMBO Mol Med 2010; 2: 504–515.
- 21 Clausen BE, Burkhardt C, Reith W, et al. Conditional gene targeting in macrophages and granulocytes using LysMcre mice. Transgenic Res 1999; 8: 265–277.
- 22 Caton ML, Smith-Raska MR, Reizis B. Notch-RBP-J signaling controls the homeostasis of CD8- dendritic cells in the spleen. J Exp Med 2007; 204: 1653–1664.
- 23 Nishida K, Yamaguchi O, Hirotani S, et al. p38alpha mitogen-activated protein kinase plays a critical role in cardiomyocyte survival but not in cardiac hypertrophic growth in response to pressure overload. Mol Cell Biol 2004; 24: 10611–10620.
- 24 Lee PP, Fitzpatrick DR, Beard C, et al. A critical role for Dnmt1 and DNA methylation in T cell development, function, and survival. Immunity 2001; 15: 763–774.
- 25 Noubade R, Milligan G, Zachary JF, et al. Histamine receptor H1 is required for TCR-mediated p38 MAPK activation and optimal IFN-gamma production in mice. J Clin Invest 2007; 117: 3507–3518.
- 26 Benjamini Y, Hochberg Y. Controlling the false discovery rate: a practical and powerful approach to multiple testing. J R Stat Soc Series B Stat Methodol 1995; 57: 289–300.
- 27 Spence RD, Voskuhl RR. Neuroprotective effects of estrogens and androgens in CNS inflammation and neurodegeneration. Front Neuroendocrinol 2012; 33: 105–115.
- 28 Segal BM. Th17 cells in autoimmune demyelinating disease. Semin Immunopathol 2010; 32: 71–77.
- 29 Rincon M, Enslen H, Raingeaud J, et al. Interferon-gamma expression by Th1 effector T cells mediated by the p38 MAP kinase signaling pathway. EMBO J 1998; 17: 2817–2829.
- 30 Chastain EM, Duncan DS, Rodgers JM, Miller SD. The role of antigen presenting cells in multiple sclerosis. Biochim Biophys Acta 2011; 1812: 265–274.
- 31 Izikson L, Klein RS, Luster AD, Weiner HL. Targeting monocyte recruitment in CNS autoimmune disease. Clin Immunol 2002; 103: 125–131.
- 32 Ponomarev ED, Shriver LP, Maresz K, Dittel BN. Microglial cell activation and proliferation precedes the onset of CNS autoimmunity. J Neurosci Res 2005; 81: 374–389.
- 33 Marta M, Meier UC, Lobell A. Regulation of autoimmune encephalomyelitis by toll-like receptors. Autoimmun Rev 2009; 8: 506–509.
- 34 Oksenberg JR, Hauser SL. Decoding multiple sclerosis. Ann Neurol 2011; 70: A5–A7.
- 35 Gourraud PA, Harbo HF, Hauser SL, Baranzini SE. The genetics of multiple sclerosis: an up-to-date review. Immunol Rev 2012; 248: 87–103.
- 36 Park JM, Greten FR, Wong A, et al. Signaling pathways and genes that inhibit pathogen-induced macrophage apoptosis—CREB and NF-kappaB as key regulators. Immunity 2005; 23: 319–329.
- 37 Kim C, Sano Y, Todorova K, et al. The kinase p38 alpha serves cell type-specific inflammatory functions in skin injury and coordinates pro- and anti-inflammatory gene expression. Nat Immunol 2008; 9: 1019–1027.
- 38 Greten FR, Arkan MC, Bollrath J, et al. NF-kappaB is a negative regulator of IL-1beta secretion as revealed by genetic and pharmacological inhibition of IKKbeta. Cell 2007; 130: 918–931.
- 39 Rosenberg GA. Matrix metalloproteinases in neuroinflammation. Glia 2002; 39: 279–291.
- 40 Carrier Y, Ma HL, Ramon HE, et al. Inter-regulation of Th17 cytokines and the IL-36 cytokines in vitro and in vivo: implications in psoriasis pathogenesis. J Invest Dermatol 2011; 131: 2428–2437.
- 41 Tortola L, Rosenwald E, Abel B, et al. Psoriasiform dermatitis is driven by IL-36-mediated DC-keratinocyte crosstalk. J Clin Invest 2012; 122: 3965–3976.
- 42 Vigne S, Palmer G, Lamacchia C, et al. IL-36R ligands are potent regulators of dendritic and T cells. Blood 2011; 118: 5813–5823.
- 43 Guan H, Fan D, Mrelashvili D, et al. MicroRNA let-7e is associated with the pathogenesis of experimental autoimmune encephalomyelitis. Eur J Immunol 2013; 43: 104–114.
- 44 Rottman JB, Slavin AJ, Silva R, et al. Leukocyte recruitment during onset of experimental allergic encephalomyelitis is CCR1 dependent. Eur J Immunol 2000; 30: 2372–2377.
- 45 Musgrave T, Benson C, Wong G, et al. The MAO inhibitor phenelzine improves functional outcomes in mice with experimental autoimmune encephalomyelitis (EAE). Brain Behav Immun 2011; 25: 1677–1688.
- 46 O'Brien M, Lonergan R, Costelloe L, et al. OAS1: a multiple sclerosis susceptibility gene that influences disease severity. Neurology 2010; 75: 411–418.
- 47 Fedetz M, Matesanz F, Caro-Maldonado A, et al. OAS1 gene haplotype confers susceptibility to multiple sclerosis. Tissue Antigens 2006; 68: 446–449.
- 48 Cagliani R, Fumagalli M, Guerini FR, et al. Identification of a new susceptibility variant for multiple sclerosis in OAS1 by population genetics analysis. Hum Genet 2012; 131: 87–97.
- 49 Rott O, Fleischer B, Cash E. Interleukin-10 prevents experimental allergic encephalomyelitis in rats. Eur J Immunol 1994; 24: 1434–1440.
- 50 Cua DJ, Sherlock J, Chen Y, et al. Interleukin-23 rather than interleukin-12 is the critical cytokine for autoimmune inflammation of the brain. Nature 2003; 421: 744–748.
- 51 Godl K, Wissing J, Kurtenbach A, et al. An efficient proteomics method to identify the cellular targets of protein kinase inhibitors. Proc Natl Acad Sci U S A 2003; 100: 15434–15439.
- 52 Spach KM, Noubade R, McElvany B, et al. A single nucleotide polymorphism in Tyk2 controls susceptibility to experimental allergic encephalomyelitis. J Immunol 2009; 182: 7776–7783.
- 53 Blankenhorn EP, Butterfield RJ, Rigby R, et al. Genetic analysis of the influence of pertussis toxin on experimental allergic encephalomyelitis susceptibility: an environmental agent can override genetic checkpoints. J Immunol 2000; 164: 3420–3425.
- 54 Matsuki T, Nakae S, Sudo K, et al. Abnormal T cell activation caused by the imbalance of the IL-1/IL-1R antagonist system is responsible for the development of experimental autoimmune encephalomyelitis. Int Immunol 2006; 18: 399–407.
- 55 Guma M, Hammaker D, Topolewski K, et al. Antiinflammatory functions of p38 in mouse models of rheumatoid arthritis: advantages of targeting upstream kinases MKK-3 or MKK-6. Arthritis Rheum 2012; 64: 2887–2895.
- 56 Clark AR, Dean JL, Saklatvala J. Post-transcriptional regulation of gene expression by mitogen-activated protein kinase p38. FEBS Lett 2003; 546: 37–44.
- 57 Schindler JF, Monahan JB, Smith WG. p38 pathway kinases as anti-inflammatory drug targets. J Dent Res 2007; 86: 800–811.
- 58 Oertelt-Prigione S. The influence of sex and gender on the immune response. Autoimmun Rev 2012; 11: A479–A485.
- 59 Imahara SD, Jelacic S, Junker CE, O'Keefe GE. The influence of gender on human innate immunity. Surgery 2005; 138: 275–282.
- 60 Sheikh-Hamad D, Gustin MC. MAP kinases and the adaptive response to hypertonicity: functional preservation from yeast to mammals. Am J Physiol Renal Physiol 2004; 287: F1102–F1110.
- 61 Cowan KJ, Storey KB. Mitogen-activated protein kinases: new signaling pathways functioning in cellular responses to environmental stress. J Exp Biol 2003; 206(pt 7): 1107–1115.
- 62 Muthusamy V, Piva TJ. The UV response of the skin: a review of the MAPK, NFkappaB and TNFalpha signal transduction pathways. Arch Dermatol Res 2010; 302: 5–17.
- 63 Orton SM, Wald L, Confavreux C, et al. Association of UV radiation with multiple sclerosis prevalence and sex ratio in France. Neurology 2011; 76: 425–431.
- 64 Norval M, McLoone P, Lesiak A, Narbutt J. The effect of chronic ultraviolet radiation on the human immune system. Photochem Photobiol 2008; 84: 19–28.
- 65 Hauser SL, Weiner HL, Che M, et al. Prevention of experimental allergic encephalomyelitis (EAE) in the SJL/J mouse by whole body ultraviolet irradiation. J Immunol 1984; 132: 1276–1281.
- 66 Becklund BR, Severson KS, Vang SV, DeLuca HF. UV radiation suppresses experimental autoimmune encephalomyelitis independent of vitamin D production. Proc Natl Acad Sci U S A 2010; 107: 6418–6423.
- 67 Kleinewietfeld M, Manzel A, Titze J, et al. Sodium chloride drives autoimmune disease by the induction of pathogenic T17 cells. Nature 2013; 496: 518–522.
- 68 Wu C, Yosef N, Thalhamer T, et al. Induction of pathogenic T17 cells by inducible salt-sensing kinase SGK1. Nature 2013; 496: 513–517.