Review Topical Sections

IL-17 signaling is regulated through intrinsic stability control of mRNA during inflammation

  • Received: 17 August 2022 Revised: 16 September 2022 Accepted: 22 September 2022 Published: 26 September 2022
  • Interleukin (IL)-17 is a proinflammatory cytokine mainly produced by immune cells, especially activated T-helper 17 cells, which contribute to chronic inflammatory and autoimmune diseases including psoriasis. Although the molecular mechanisms of transcription in IL-17-mediated signaling pathways are well established, post-transcriptional control remains to be elucidated. Notably, IL-17 regulates post-transcriptional modifications, which induce elevated levels of target inflammatory mRNAs. Regnase-1, an endoribonuclease and deubiquitinase, post-transcriptionally downregulates various IL-17-driven signaling pathways, including mRNA stability. The ACT1-TBK1/IKKϵ pathway and ARID5A were induced and activated by IL-17-stimulation, leading to the inhibition of inflammatory mRNA degradation by Regnase-1. In this review, we focus on IL-17-mediated mRNA stabilization of psoriasis-related IκB-ζ and provide novel therapeutic strategies for the treatment of Th17-mediated inflammation and autoimmunity.

    Citation: Ryuta Muromoto, Kenji Oritani, Tadashi Matsuda. IL-17 signaling is regulated through intrinsic stability control of mRNA during inflammation[J]. AIMS Allergy and Immunology, 2022, 6(3): 188-199. doi: 10.3934/Allergy.2022014

    Related Papers:

  • Interleukin (IL)-17 is a proinflammatory cytokine mainly produced by immune cells, especially activated T-helper 17 cells, which contribute to chronic inflammatory and autoimmune diseases including psoriasis. Although the molecular mechanisms of transcription in IL-17-mediated signaling pathways are well established, post-transcriptional control remains to be elucidated. Notably, IL-17 regulates post-transcriptional modifications, which induce elevated levels of target inflammatory mRNAs. Regnase-1, an endoribonuclease and deubiquitinase, post-transcriptionally downregulates various IL-17-driven signaling pathways, including mRNA stability. The ACT1-TBK1/IKKϵ pathway and ARID5A were induced and activated by IL-17-stimulation, leading to the inhibition of inflammatory mRNA degradation by Regnase-1. In this review, we focus on IL-17-mediated mRNA stabilization of psoriasis-related IκB-ζ and provide novel therapeutic strategies for the treatment of Th17-mediated inflammation and autoimmunity.



    加载中

    Acknowledgments



    The authors thank Editage (www.editage.com) for English language editing. This study was supported in part by Grant-in-Aid for scientific research 20K07010 (R. M.) and 19H03364 (T.M.) from Ministry of Education, Culture, Sports, Science and Technology of Japan.

    Conflict of interest



    All authors declare no conflicts of interest in this paper.

    [1] Mosmann TR, Coffman RL (1989) TH1 and TH2 cells: different patterns of lymphokine secretion lead to different functional properties. Annu Rev Immunol 7: 145-173. https://doi.org/10.1146/annurev.iy.07.040189.001045
    [2] Korn T, Bettelli E, Oukka M, et al. (2009) IL-17 and Th17 Cells. Annu Rev Immunol 27: 485-517. https://doi.org/10.1146/annurev.immunol.021908.132710
    [3] Chung SH, Ye XQ, Iwakura Y, et al. (2021) Interleukin-17 family members in health and disease. Int Immunol 33: 723-729. https://doi.org/10.1093/intimm/dxab075
    [4] Mills KHG (2022) IL-17 and IL-17-producing cells in protection versus pathology. Nat Rev Immunol 2022: 1-17. https://doi.org/10.1038/s41577-022-00746-9
    [5] Lee GR (2018) The balance of Th17 versus Treg cells in autoimmunity. Int J Mol Sci 19: 730. https://doi.org/10.3390/ijms19030730
    [6] Milovanovic J, Arsenijevic A, Stojanovic B, et al. (2020) Interleukin-17 in chronic inflammatory neurological diseases. Front Immunol 11: 947. https://doi.org/10.3389/fimmu.2020.00947
    [7] von Stebut E, Boehncke WH, Ghoreschi K, et al. (2020) IL-17A in psoriasis and beyond: Cardiovascular and metabolic implications. Front Immunol 10: 3096. https://doi.org/10.3389/fimmu.2019.03096
    [8] Akira S, Maeda K (2021) Control of RNA stability in immunity. Annu Rev Immunol 39: 481-509. https://doi.org/10.1146/annurev-immunol-101819-075147
    [9] Yoshinaga M, Takeuchi O (2019) Post-transcriptional control of immune responses and its potential application. Clin Transl Immunol 8: e1063. https://doi.org/10.1002/cti2.1063
    [10] Qian Y, Liu C, Hartupee J, et al. (2007) The adaptor Act1 is required for interleukin 17-dependent signaling associated with autoimmune and inflammatory disease. Nat Immunol 8: 247-256. https://doi.org/10.1038/ni1439
    [11] Onishi RM, Gaffen SL (2010) Interleukin-17 and its target genes: mechanisms of interleukin-17 function in disease. Immunology 129: 311-321. https://doi.org/10.1111/j.1365-2567.2009.03240.x
    [12] Tohyama M, Hanakawa Y, Shirakata Y, et al. (2009) IL-17 and IL-22 mediate IL-20 subfamily cytokine production in cultured keratinocytes via increased IL-22 receptor expression. Eur J Immunol 39: 2779-2788. https://doi.org/10.1002/eji.200939473
    [13] Anderson P (2009) Intrinsic mRNA stability helps compose the inflammatory symphony. Nat Immunol 10: 233-234. https://doi.org/10.1038/ni0309-233
    [14] Anderson P (2010) Post-transcriptional regulons coordinate the initiation and resolution of inflammation. Nat Rev Immunol 10: 24-35. https://doi.org/10.1038/nri2685
    [15] Masuda K, Ripley B, Nishimura R, et al. (2013) Arid5a controls IL-6 mRNA stability, which contributes to elevation of IL-6 level in vivo. P Natl Acad Sci USA 110: 9409-9414. https://doi.org/10.1073/pnas.1307419110
    [16] Matsushita K, Takeuchi O, Standley DM, et al. (2009) Zc3h12a is an RNase essential for controlling immune responses by regulating mRNA decay. Nature 458: 1185-1190. https://doi.org/10.1038/nature07924
    [17] Uehata T, Iwasaki H, Vandenbon A, et al. (2013) Malt1-induced cleavage of regnase-1 in CD4(+) helper T cells regulates immune activation. Cell 153: 1036-1049. https://doi.org/10.1016/j.cell.2013.04.034
    [18] Garg AV, Amatya N, Chen K, et al. (2015) MCPIP1 Endoribonuclease activity negatively regulates interleukin-17-mediated signaling and inflammation. Immunity 43: 475-487. https://doi.org/10.1016/j.immuni.2015.07.021
    [19] Amatya N, Childs EE, Cruz JA, et al. (2018) IL-17 integrates multiple self-reinforcing, feed-forward mechanisms through the RNA binding protein Arid5a. Sci Signal 11: eaat4617. https://doi.org/10.1126/scisignal.aat4617
    [20] Tanaka H, Arima Y, Kamimura D, et al. (2019) Phosphorylation-dependent Regnase-1 release from endoplasmic reticulum is critical in IL-17 response. J Exp Med 216: 1431-1449. https://doi.org/10.1084/jem.20181078
    [21] Bulek K, Liu C, Swaidani S, et al. (2011) The inducible kinase IKKi is required for IL-17-dependent signaling associated with neutrophilia and pulmonary inflammation. Nat Immunol 12: .844-852. https://doi.org/10.1038/ni.2080
    [22] Qu F, Gao H, Zhu S, et al. (2012) TRAF6-dependent Act1 phosphorylation by the IκB kinase-related kinases suppresses interleukin-17-induced NF-κB activation. Mol Cell Biol 32: 3925-3937. https://doi.org/10.1128/MCB.00268-12
    [23] Muromoto R, Hirao T, Tawa K, et al. (2016) IL-17A plays a central role in the expression of psoriasis signature genes through the induction of IκB-ζ in keratinocytes. Int Immunol 28: 443-452. https://doi.org/10.1093/intimm/dxw011
    [24] Willems M, Dubois N, Musumeci L (2016) IκBζ: an emerging player in cancer. Oncotarget 7: 66310-66322. https://doi.org/10.18632/oncotarget.11624
    [25] Bertelsen T, Ljungberg C, Kjellerup RB, et al. (2017) IL-17F regulates psoriasis-associated genes through IκBζ. Exp Dermatol 26: 234-241. https://doi.org/10.1111/exd.13182
    [26] Müller A, Hennig A, Lorscheid S, et al. (2018) IκBζ is a key transcriptional regulator of IL-36-driven psoriasis-related gene expression in keratinocytes. P Natl Acad Sci USA 115: 10088-10093. https://doi.org/10.1073/pnas.1801377115
    [27] Yamazaki S, Muta T, Matsuo S, et al. (2005) Stimulus-specific induction of a novel nuclear factor-κB regulator, IκB-ζ, via toll/interleukin-1 receptor is mediated by mRNA stabilization. J Biol Chem 280: 1678-1687. https://doi.org/10.1074/jbc.M409983200
    [28] Okuma A, Hoshino K, Ohba T, et al. (2013) Enhanced apoptosis by disruption of the STAT3-IκB-ζ signaling pathway in epithelial cells induces Sjögren's syndrome-like autoimmune disease. Immunity 38: 450-460. https://doi.org/10.1016/j.immuni.2012.11.016
    [29] Muromoto R, Tawa K, Ohgakiuchi Y, et al. IκB-ζ expression requires both TYK2/STAT3 activity and IL-17-regulated mRNA stabilization. Immunohorizons 3: 172-185. https://doi.org/10.4049/immunohorizons.1900023
    [30] Ohgakiuchi Y, Saino Y, Muromoto R, et al. (2020) Dimethyl fumarate dampens IL-17-ACT1-TBK1 axis-mediated phosphorylation of Regnase-1 and suppresses IL-17-induced IκB-ζ expression. Biochem Biophys Res Commun 521: 957-963. https://doi.org/10.1016/j.bbrc.2019.11.036
    [31] Nestle FO, Kaplan DH, Barker J (2009) Psoriasis. N Engl J Med 361: 496-509. https://doi.org/10.1056/NEJMra0804595
    [32] Christophers E, Metzler G, Röcken M (2014) Bimodal immune activation in psoriasis. Brit J Dermatol 170: 59-65. https://doi.org/10.1111/bjd.12631
    [33] Lowes MA, Suárez-Fariñas M, Krueger JG (2014) Immunology of psoriasis. Annu Rev Immunol 32: 227-255. https://doi.org/10.1146/annurev-immunol-032713-120225
    [34] Lynde CW, Poulin Y, Vender R, et al. (2014) Interleukin 17A: toward a new understanding of psoriasis pathogenesis. J Am Acad Dermatol 71: 141-150. https://doi.org/10.1016/j.jaad.2013.12.036
    [35] Baliwag J, Barnes DH, Johnston A (2015) Cytokines in psoriasis. Cytokine 73: 342-350. https://doi.org/10.1016/j.cyto.2014.12.014
    [36] Lorscheid S, Müller A, Löffler J, et al. (2019) Keratinocyte-derived IκBζ drives psoriasis and associated systemic inflammation. JCI Insight 4: e130835. https://doi.org/10.1172/jci.insight.130835
    [37] Mandal A, Kumbhojkar N, Reilly C, et al. (2020) Treatment of psoriasis with NFKBIZ siRNA using topical ionic liquid formulations. Sci Adv 6: eabb6049. https://doi.org/10.1126/sciadv.abb6049
    [38] Schwartz DM, Kanno Y, Villarino A, et al. (2017) JAK inhibition as a therapeutic strategy for immune and inflammatory diseases. Nat Rev Drug Discov 16: 843-862. https://doi.org/10.1038/nrd.2017.201
    [39] Muromoto R, Oritani K, Matsuda T (2022) Current understanding of the role of tyrosine kinase 2 signaling in immune responses. World J Biol Chem 13: 1-14. https://doi.org/10.4331/wjbc.v13.i1.1
    [40] Muromoto R, Shimoda K, Oritani K, et al. (2021) Therapeutic advantage of Tyk2 inhibition for Treating autoimmune and chronic inflammatory diseases. Biol Pharm Bull 44: 1585-1592. https://doi.org/10.1248/bpb.b21-00609
    [41] Burke JR, Cheng L, Gillooly KM, et al. (2019) Autoimmune pathways in mice and humans are blocked by pharmacological stabilization of the TYK2 pseudokinase domain. Sci Transl Med 11: eaaw1736. https://doi.org/10.1126/scitranslmed.aaw1736
    [42] Chang Y, Xu S, Ding K (2019) Tyrosine kinase 2 (TYK2) allosteric inhibitors to treat autoimmune diseases. J Med Chem 62: 8951-8952. https://doi.org/10.1021/acs.jmedchem.9b01612
    [43] Sadeghi A, Tahmasebi S, Mahmood A, et al. (2021) Th17 and Treg cells function in SARS-CoV2 patients compared with healthy controls. J Cell Physiol 236: 2829-2839. https://doi.org/10.1002/jcp.30047
    [44] Leija-Martínez JJ, Huang F, Del-Río-Navarro BE, et al. (2020) IL-17A and TNF-α as potential biomarkers for acute respiratory distress syndrome and mortality in patients with obesity and COVID-19. Med Hypotheses 144: 109935. https://doi.org/10.1016/j.mehy.2020.109935
    [45] Hasan MZ, Islam S, Matsumoto K, et al. (2021) SARS-CoV-2 infection initiates interleukin-17-enriched transcriptional response in different cells from multiple organs. Sci Rep 11: 16814. https://doi.org/10.1038/s41598-021-96110-3
    [46] Maslennikov R, Ivashkin V, Vasilieva E, et al. (2021) Interleukin 17 antagonist netakimab is effective and safe in the new coronavirus infection (COVID-19). Eur Cytokine Netw 32: 8-14. https://doi.org/10.1684/ecn.2021.0463
    [47] Avdeev SN, Trushenko NV, Tsareva NA, et al. (2021) Anti-IL-17 monoclonal antibodies in hospitalized patients with severe COVID-19: a pilot study. Cytokine 146: 155627. https://doi.org/10.1016/j.cyto.2021.155627
  • Reader Comments
  • © 2022 the Author(s), licensee AIMS Press. This is an open access article distributed under the terms of the Creative Commons Attribution License (http://creativecommons.org/licenses/by/4.0)
通讯作者: 陈斌, bchen63@163.com
  • 1. 

    沈阳化工大学材料科学与工程学院 沈阳 110142

  1. 本站搜索
  2. 百度学术搜索
  3. 万方数据库搜索
  4. CNKI搜索

Metrics

Article views(1225) PDF downloads(107) Cited by(0)

Article outline

Figures and Tables

Figures(2)

Other Articles By Authors

/

DownLoad:  Full-Size Img  PowerPoint
Return
Return

Catalog