DOI QR코드

DOI QR Code

GRIM-19 Ameliorates Multiple Sclerosis in a Mouse Model of Experimental Autoimmune Encephalomyelitis with Reciprocal Regulation of IFNγ/Th1 and IL-17A/Th17 Cells

  • Jeonghyeon Moon (Laboratory of Immune Network, Conversant Research Consortium in Immunologic Disease, College of Medicine, The Catholic University of Korea) ;
  • Seung Hoon Lee (Rheumatism Research Center, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea) ;
  • Seon-yeong Lee (Rheumatism Research Center, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea) ;
  • Jaeyoon Ryu (Rheumatism Research Center, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea) ;
  • Jooyeon Jhun (Rheumatism Research Center, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea) ;
  • JeongWon Choi (Rheumatism Research Center, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea) ;
  • Gyoung Nyun Kim (College of Medicine, The Catholic University of Korea) ;
  • Sangho Roh (Cellular Reprogramming and Embryo Biotechnology Laboratory, Dental Research Institute, BK21 PLUS Dental Life Science, Seoul National University School of Dentistry) ;
  • Sung-Hwan Park (Division of Rheumatology, Department of Internal Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea) ;
  • Mi-La Cho (Laboratory of Immune Network, Conversant Research Consortium in Immunologic Disease, College of Medicine, The Catholic University of Korea)
  • Received : 2020.07.22
  • Accepted : 2020.09.23
  • Published : 2020.10.31

Abstract

The protein encoded by the Gene Associated with Retinoid-Interferon-Induced Mortality-19 (GRIM-19) is located in the mitochondrial inner membrane and is homologous to the NADH dehydrogenase 1-alpha subcomplex subunit 13 of the electron transport chain. Multiple sclerosis (MS) is a demyelinating disease that damages the brain and spinal cord. Although both the cause and mechanism of MS progression remain unclear, it is accepted that an immune disorder is involved. We explored whether GRIM-19 ameliorated MS by increasing the levels of inflammatory cytokines and immune cells; we used a mouse model of experimental autoimmune encephalomyelitis (EAE) to this end. Six-to-eight-week-old male C57BL/6, IFNγ-knockout (KO), and GRIM-19 transgenic mice were used; EAE was induced in all strains. A GRIM-19 overexpression vector (GRIM19 OVN) was electrophoretically injected intravenously. The levels of Th1 and Th17 cells were measured via flow cytometry, immunofluorescence, and immunohistochemical analysis. IL-17A and IFNγ expression levels were assessed via ELISA and quantitative PCR. IL-17A expression decreased and IFNγ expression increased in EAE mice that received injections of the GRIM-19 OVN. GRIM19 transgenic mice expressed more IFNγ than did wild-type mice; this inhibited EAE development. However, the effect of GRIM-19 overexpression on the EAE of IFNγ-KO mice did not differ from that of the empty vector. GRIM-19 expression was therapeutic for EAE mice, elevating the IFNγ level. GRIM-19 regulated the Th17/Treg cell balance.

Keywords

Acknowledgement

This research was supported by a grant of the Korea Health Technology R&D Project through the Korea Health Industry Development Institute (KHIDI), funded by the Ministry of Health & Welfare, Republic of Korea (grant number HI20C1496) and a grant of the Korea Health Technology R&D Project through the Korea Health Industry Development Institute (KHIDI), funded by the Ministry of Health & Welfare, Republic of Korea (grant number: HI15C3062).

References

  1. Huang G, Lu H, Hao A, Ng DC, Ponniah S, Guo K, Lufei C, Zeng Q, Cao X. GRIM-19, a cell death regulatory protein, is essential for assembly and function of mitochondrial complex I. Mol Cell Biol 2004;24:8447-8456.
  2. Tian B, Zhao Y, Liang T, Ye X, Li Z, Yan D, Fu Q, Li Y. Curcumin inhibits urothelial tumor development by suppressing IGF2 and IGF2-mediated PI3K/AKT/mTOR signaling pathway. J Drug Target 2017;25:626-636.
  3. Lufei C, Ma J, Huang G, Zhang T, Novotny-Diermayr V, Ong CT, Cao X. GRIM-19, a death-regulatory gene product, suppresses Stat3 activity via functional interaction. EMBO J 2003;22:1325-1335.
  4. Zhang J, Yang J, Roy SK, Tininini S, Hu J, Bromberg JF, Poli V, Stark GR, Kalvakolanu DV. The cell death regulator GRIM-19 is an inhibitor of signal transducer and activator of transcription 3. Proc Natl Acad Sci U S A 2003;100:9342-9347.
  5. Kalvakolanu DV, Nallar SC, Kalakonda S. Cytokine-induced tumor suppressors: a GRIM story. Cytokine 2010;52:128-142.
  6. Kong D, Zhao L, Du Y, He P, Zou Y, Yang L, Sun L, Wang H, Xu D, Meng X, et al. Overexpression of GRIM19, a mitochondrial respiratory chain complex I protein, suppresses hepatocellular carcinoma growth. Int J Clin Exp Pathol 2014;7:7497-7507.
  7. Nallar SC, Kalakonda S, Sun P, Kalvakolanu DV. Grim-19: A double-edged sword that regulates anti-tumor and innate immune responses. Transl Oncogenomics 2008.3:67-79.
  8. Chen W, Liu Q, Fu B, Liu K, Jiang W. Overexpression of GRIM-19 accelerates radiation-induced osteosarcoma cells apoptosis by p53 stabilization. Life Sci 2018;208:232-238.
  9. Li F, Ren W, Zhao Y, Fu Z, Ji Y, Zhu Y, Qin C. Downregulation of GRIM-19 is associated with hyperactivation of p-STAT3 in hepatocellular carcinoma. Med Oncol 2012;29:3046-3054.
  10. Wang T, Yan XB, Zhao JJ, Ye J, Jiang ZF, Wu DR, Xiao WH, Liu RY. Gene associated with retinoid-interferon-induced mortality-19 suppresses growth of lung adenocarcinoma tumor in vitro and in vivo. Lung Cancer 2011;72:287-293.
  11. Zhou Y, Wei Y, Zhu J, Wang Q, Bao L, Ma Y, Chen Y, Feng D, Zhang A, Sun J, et al. GRIM-19 disrupts E6/E6AP complex to rescue p53 and induce apoptosis in cervical cancers. PLoS One 2011;6:e22065.
  12. Lin H, Shen Z, Liu H, Yang M, Lin J, Luo L, Liu L, Chen H. Upregulation of GRIM-19 augments the sensitivity of prostate cancer cells to docetaxel by targeting Rad23b. Clin Exp Pharmacol Physiol 2020;47:76-84.
  13. Chen H, Deng X, Yang Y, Shen Y, Chao L, Wen Y, Sun Y. Expression of GRIM-19 in missed abortion and possible pathogenesis. Fertil Steril 2015;103:138-46.e3.
  14. Fjaer S, Bo L, Myhr KM, Torkildsen O, Wergeland S. Magnetization transfer ratio does not correlate to myelin content in the brain in the MOG-EAE mouse model. Neurochem Int 2015;83-84:28-40.
  15. Galea I, Ward-Abel N, Heesen C. Relapse in multiple sclerosis. BMJ 2015;350:h1765.
  16. Lorscheider J, Buzzard K, Jokubaitis V, Spelman T, Havrdova E, Horakova D, Trojano M, Izquierdo G, Girard M, Duquette P, et al. Defining secondary progressive multiple sclerosis. Brain 2016;139:2395-2405.
  17. Owens B. Multiple sclerosis. Nature 2016;540:S1.
  18. Bittner S, Afzali AM, Wiendl H, Meuth SG. Myelin oligodendrocyte glycoprotein (MOG35-55) induced experimental autoimmune encephalomyelitis (EAE) in C57BL/6 mice. J Vis Exp 2014:51275.
  19. Alharbi FM. Update in vitamin D and multiple sclerosis. Neurosciences 2015;20:329-335.
  20. Rolak LA. Multiple sclerosis: it's not the disease you thought it was. Clin Med Res 2003;1:57-60.
  21. Faguy K. Multiple sclerosis: an update. Radiol Technol 2016;87:529-550.
  22. Garg N, Smith TW. An update on immunopathogenesis, diagnosis, and treatment of multiple sclerosis. Brain Behav 2015;5:e00362.
  23. Yadav SK, Mindur JE, Ito K, Dhib-Jalbut S. Advances in the immunopathogenesis of multiple sclerosis. Curr Opin Neurol 2015;28:206-219.
  24. Darlington PJ, Boivin MN, Renoux C, Francois M, Galipeau J, Freedman MS, Atkins HL, Cohen JA, Solchaga L, Bar-Or A. Reciprocal Th1 and Th17 regulation by mesenchymal stem cells: implication for multiple sclerosis. Ann Neurol 2010;68:540-545.
  25. Quinones-Aguilar S, Sauri-Suarez S, Alcaraz-Estrada SL, Garcia S. Progressive multifocal leukoencephalopathy associated to treatment with natalizumab in Mexican patient multiple sclerosis. Case report, analysis and update. Neuroendocrinol Lett 2019;40:222-226.
  26. Procaccini C, De Rosa V, Pucino V, Formisano L, Matarese G. Animal models of multiple sclerosis. Eur J Pharmacol 2015;759:182-191.
  27. Kuerten S, Javeri S, Tary-Lehmann M, Lehmann PV, Angelov DN. Fundamental differences in the dynamics of CNS lesion development and composition in MP4- and MOG peptide 35-55-induced experimental autoimmune encephalomyelitis. Clin Immunol 2008;129:256-267.
  28. Linker RA, Lee DH. Models of autoimmune demyelination in the central nervous system: on the way to translational medicine. Exp Transl Stroke Med 2009;1:5.
  29. Bowles AC, Scruggs BA, Bunnell BA. Mesenchymal stem cell-based therapy in a mouse model of experimental autoimmune encephalomyelitis (EAE). Methods Mol Biol 2014;1213:303-319. 
  30. Aranami T, Yamamura T. Th17 Cells and autoimmune encephalomyelitis (EAE/MS). Allergol Int 2008;57:115-120.
  31. Baker D, Amor S. Mouse models of multiple sclerosis: lost in translation? Curr Pharm Des 2015;21:2440-2452.
  32. Pettinelli CB, McFarlin DE. Adoptive transfer of experimental allergic encephalomyelitis in SJL/J mice after in vitro activation of lymph node cells by myelin basic protein: requirement for Lyt 1+ 2- T lymphocytes. J Immunol 1981;127:1420-1423.
  33. Ando DG, Clayton J, Kono D, Urban JL, Sercarz EE. Encephalitogenic T cells in the B10.PL model of experimental allergic encephalomyelitis (EAE) are of the Th-1 lymphokine subtype. Cell Immunol 1989;124:132-143.
  34. Hofstetter HH, Ibrahim SM, Koczan D, Kruse N, Weishaupt A, Toyka KV, Gold R. Therapeutic efficacy of IL-17 neutralization in murine experimental autoimmune encephalomyelitis. Cell Immunol 2005;237:123-130.
  35. Ponomarev ED, Shriver LP, Maresz K, Pedras-Vasconcelos J, Verthelyi D, Dittel BN. GM-CSF production by autoreactive T cells is required for the activation of microglial cells and the onset of experimental autoimmune encephalomyelitis. J Immunol 2007;178:39-48.
  36. El-Behi M, Ciric B, Dai H, Yan Y, Cullimore M, Safavi F, Zhang GX, Dittel BN, Rostami A. The encephalitogenicity of TH17 cells is dependent on IL-1- and IL-23-induced production of the cytokine GM-CSF. Nat Immunol 2011;12:568-575.
  37. Cummings M, Arumanayagam AC, Zhao P, Kannanganat S, Stuve O, Karandikar NJ, Eagar TN. Presenilin1 regulates Th1 and Th17 effector responses but is not required for experimental autoimmune encephalomyelitis. PLoS One 2018;13:e0200752.
  38. Giacoppo S, Pollastro F, Grassi G, Bramanti P, Mazzon E. Target regulation of PI3K/Akt/mTOR pathway by cannabidiol in treatment of experimental multiple sclerosis. Fitoterapia 2017;116:77-84.
  39. Hamana A, Takahashi Y, Tanioka A, Nishikawa M, Takakura Y. Amelioration of experimental autoimmune encephalomyelitis in mice by interferon-beta gene therapy, using a long-term expression plasmid vector. Mol Pharm 2017;14:1212-1217.
  40. Hou H, Miao J, Cao R, Han M, Sun Y, Liu X, Guo L. Rapamycin ameliorates experimental autoimmune encephalomyelitis by suppressing the mTOR-STAT3 pathway. Neurochem Res 2017;42:2831-2840.
  41. Suda T, Liu D. Hydrodynamic gene delivery: its principles and applications. Mol Ther 2007;15:2063-2069.
  42. Angell JE, Lindner DJ, Shapiro PS, Hofmann ER, Kalvakolanu DV. Identification of GRIM-19, a novel cell death-regulatory gene induced by the interferon-β and retinoic acid combination, using a genetic approach. J Biol Chem 2000;275:33416-33426.
  43. Lu H, Cao X. GRIM-19 is essential for maintenance of mitochondrial membrane potential. Mol Biol Cell 2008;19:1893-1902.
  44. Yamamoto-Furusho JK, Barnich N, Hisamatsu T, Podolsky DK. MDP-NOD2 stimulation induces HNP-1 secretion, which contributes to NOD2 antibacterial function. Inflamm Bowel Dis 2010;16:736-742.
  45. Strober W, Murray PJ, Kitani A, Watanabe T. Signalling pathways and molecular interactions of NOD1 and NOD2. Nat Rev Immunol 2006;6:9-20.
  46. Moon YM, Lee J, Lee SY, Her YM, Ryu JG, Kim EK, Son HJ, Kwok SK, Ju JH, Yang CW, et al. Gene associated with retinoid-interferon-induced mortality 19 attenuates murine autoimmune arthritis by regulation of Th17 and Treg cells. Arthritis Rheumatol 2014;66:569-578.
  47. Kim JK, Lee SH, Lee SY, Kim EK, Kwon JE, Seo HB, Lee HH, Lee BI, Park SH, Cho ML. Grim19 attenuates DSS induced colitis in an animal model. PLoS One 2016;11:e0155853. 
  48. Park MJ, Lee SH, Lee SH, Kim EK, Lee EJ, Moon YM, La Cho M. GRIM19 ameliorates acute graft-versus-host disease (GVHD) by modulating Th17 and Treg cell balance through down-regulation of STAT3 and NF-AT activation. J Transl Med 2016;14:206.
  49. Lee J, Lee J, Park MK, Lim MA, Park EM, Kim EK, Yang EJ, Lee SY, Jhun JY, Park SH, et al. Interferon gamma suppresses collagen-induced arthritis by regulation of Th17 through the induction of indoleamine-2,3-deoxygenase. PLoS One 2013;8:e60900.
  50. Lee SH, Kwon JY, Kim SY, Jung K, Cho ML. Interferon-gamma regulates inflammatory cell death by targeting necroptosis in experimental autoimmune arthritis. Sci Rep 2017;7:10133.
  51. Arellano G, Ottum PA, Reyes LI, Burgos PI, Naves R. Stage-specific role of interferon-gamma in experimental autoimmune encephalomyelitis and multiple sclerosis. Front Immunol 2015;6:492.
  52. Miller NM, Wang J, Tan Y, Dittel BN. Anti-inflammatory mechanisms of IFN-γ studied in experimental autoimmune encephalomyelitis reveal neutrophils as a potential target in multiple sclerosis. Front Neurosci 2015;9:287.
  53. Sosa RA, Murphey C, Robinson RR, Forsthuber TG. IFN-γ ameliorates autoimmune encephalomyelitis by limiting myelin lipid peroxidation. Proc Natl Acad Sci U S A 2015;112:E5038-E5047.
  54. Naves R, Singh SP, Cashman KS, Rowse AL, Axtell RC, Steinman L, Mountz JD, Steele C, De Sarno P, Raman C. The interdependent, overlapping, and differential roles of type I and II IFNs in the pathogenesis of experimental autoimmune encephalomyelitis. J Immunol 2013;191:2967-2977.
  55. Kalakonda S, Nallar SC, Jaber S, Keay SK, Rorke E, Munivenkatappa R, Lindner DJ, Fiskum GM, Kalvakolanu DV. Monoallelic loss of tumor suppressor GRIM-19 promotes tumorigenesis in mice. Proc Natl Acad Sci U S A 2013;110:E4213-E4222.
  56. Kalakonda S, Nallar SC, Gong P, Lindner DJ, Goldblum SE, Reddy SP, Kalvakolanu DV. Tumor suppressive protein gene associated with retinoid-interferon-induced mortality (GRIM)-19 inhibits src-induced oncogenic transformation at multiple levels. Am J Pathol 2007;171:1352-1368.
  57. Nallar SC, Kalvakolanu DV. GRIM-19: a master regulator of cytokine induced tumor suppression, metastasis and energy metabolism. Cytokine Growth Factor Rev 2017;33:1-18.
  58. Song J, Shi W, Wang W, Zhang Y, Zheng S. Grim-19 expressed by recombinant adenovirus for esophageal neoplasm target therapy. Mol Med Rep 2018;17:6667-6674.