985
Views
0
CrossRef citations to date
0
Altmetric
Original Research

Interferon regulatory factor 4 plays a pivotal role in the development of aGVHD-associated colitis

, , , , , , , , , , , , , , , , , & ORCID Icon show all
Article: 2296712 | Received 30 May 2023, Accepted 14 Dec 2023, Published online: 27 Dec 2023

References

  • Wingard JR, Majhail NS, Brazauskas R, Wang Z, Sobocinski KA, Jacobsohn D, Sorror ML, Horowitz MM, Bolwell B, Rizzo JD, et al. Long-term survival and late deaths after allogeneic hematopoietic cell transplantation. J Clin Oncol. 2011;29(16):2230–12. doi: 10.1200/JCO.2010.33.7212.
  • Ayuk F, Veit R, Zabelina T, Bussmann L, Christopeit M, Alchalby H, Wolschke C, Lellek H, Bacher U, Zander AR, et al. 2015. Prognostic factors for survival of patients with newly diagnosed chronic GVHD according to NIH criteria. Ann Hematol. 94(10):1727–1732. doi: 10.1007/s00277-015-2452-6.
  • Carnevale-Schianca F, Leisenring W, Martin PJ, Furlong T, Schoch G, Anasetti C, Appelbaum FR, Carpenter PA, Deeg HJ, Kiem H-P, et al. Longitudinal assessment of morbidity and acute graft-versus-host disease after allogeneic hematopoietic cell transplantation: retrospective analysis of a multicenter phase III study. Biol Blood Marrow Transplant. 2009;15(6):749–756. doi: 10.1016/j.bbmt.2009.03.009.
  • Jagasia M, Arora M, Flowers MED, Chao NJ, McCarthy PL, Cutler CS, Urbano-Ispizua A, Pavletic SZ, Haagenson MD, Zhang M-J, et al. 2012. Risk factors for acute GVHD and survival after hematopoietic cell transplantation. Blood. 119(1):296–307. doi: 10.1182/blood-2011-06-364265.
  • Inagaki J, Moritake H, Nishikawa T, Hyakuna N, Okada M, Suenobu S-I, Nagai K, Honda Y, Shimomura M, Fukano R, et al. 2015. Long-term morbidity and mortality in children with chronic graft-versus-host disease classified by National Institutes of Health Consensus criteria after allogeneic hematopoietic stem cell transplantation. Biol Blood Marrow Transplant. 21(11):1973–1980. doi: 10.1016/j.bbmt.2015.07.025.
  • Martin PJ, Schoch G, Fisher L, Byers V, Anasetti C, Appelbaum FR, Beatty PG, Doney K, McDonald GB, Sanders JE. A retrospective analysis of therapy for acute graft-versus-host disease: initial treatment. Blood. 1990;76(8):1464–1472. doi: 10.1182/blood.V76.8.1464.1464.
  • Yu Y, Wang D, Liu C, Kaosaard K, Semple K, Anasetti C, Yu X-Z. Prevention of GVHD while sparing GVL effect by targeting Th1 and Th17 transcription factor T-bet and RORγt in mice. Blood. 2011;118(18):5011–5020. doi: 10.1182/blood-2011-03-340315.
  • Yi T, Chen Y, Wang L, Du G, Huang D, Zhao D, Johnston H, Young J, Todorov I, Umetsu DT, et al. 2009. Reciprocal differentiation and tissue-specific pathogenesis of Th1, Th2, and Th17 cells in graft-versus-host disease. Blood. 114(14):3101–3112. doi: 10.1182/blood-2009-05-219402.
  • Nikolic B, Lee S, Bronson RT, Grusby MJ, Sykes M. Th1 and Th2 mediate acute graft-versus-host disease, each with distinct end-organ targets. J Clin Invest. 2000;105(9):1289–1298. doi: 10.1172/JCI7894.
  • Blazar BR, Taylor PA, Panoskaltsis-Mortari A, Vallera DA. Rapamycin inhibits the generation of graft-versus-host disease- and graft-versus-leukemia-causing T cells by interfering with the production of Th1 or Th1 cytotoxic cytokines. J Immunol. 1998;160(11):5355–5365. doi: 10.4049/jimmunol.160.11.5355.
  • Das H, Imoto S, Murayama T, Mizuno I, Sugimoto T, Taniguchi R, Toda K, Isobe T, Nakagawa T, Nishimura R, et al. 2001. Kinetic analysis of cytokine gene expression in patients with GVHD after donor lymphocyte infusion. Bone Marrow Transplant. 27(4):373–380. doi: 10.1038/sj.bmt.1702799.
  • Hoffmann P, Ermann J, Edinger M, Fathman CG, Strober S. Donor-type CD4(+)CD25(+) regulatory T cells suppress lethal acute graft-versus-host disease after allogeneic bone marrow transplantation. J Exp Med. 2002;196(3):389–399. doi: 10.1084/jem.20020399.
  • Semple K, Yu Y, Wang D, Anasetti C, Yu X-Z. Efficient and selective prevention of GVHD by antigen-specific induced Tregs via linked-suppression in mice. Biol Blood Marrow Transplant. 2011;17(3):309–318. doi: 10.1016/j.bbmt.2010.12.710.
  • Hippen KL, Merkel SC, Schirm DK, Sieben CM, Sumstad D, Kadidlo DM, McKenna DH, Bromberg JS, Levine BL, Riley JL, et al. Massive ex vivo expansion of human natural regulatory T cells (T(regs)) with minimal loss of in vivo functional activity. Sci Transl Med. 2011;3(83):83ra41. doi: 10.1126/scitranslmed.3001809.
  • Campe J, Ullrich E. T helper cell lineage-defining transcription factors: potent targets for specific GVHD Therapy? Front Immunol. 2021;12:806529. doi: 10.3389/fimmu.2021.806529.
  • Krenger W, Snyder KM, Byon JC, Falzarano G, Ferrara JL. Polarized type 2 alloreactive CD4+ and CD8+ donor T cells fail to induce experimental acute graft-versus-host disease. J Immunol. 1995;155(2):585–593. doi: 10.4049/jimmunol.155.2.585.
  • Pan L, Delmonte J, Jalonen CK, Ferrara JL. Pretreatment of donor mice with granulocyte colony-stimulating factor polarizes donor T lymphocytes toward type-2 cytokine production and reduces severity of experimental graft-versus-host disease. Blood. 1995;86(12):4422–4429. doi: 10.1182/blood.V86.12.4422.bloodjournal86124422.
  • Tawara I, Maeda Y, Sun Y, Lowler KP, Liu C, Toubai T, McKenzie ANJ, Reddy P. Combined Th2 cytokine deficiency in donor T cells aggravates experimental acute graft-vs-host disease. Exp Hematol. 2008;36(8):988–996. doi: 10.1016/j.exphem.2008.02.010.
  • Fulton LM, Carlson MJ, Coghill JM, Ott LE, West ML, Panoskaltsis-Mortari A, Littman DR, Blazar BR, Serody JS. Attenuation of acute graft-versus-host disease in the absence of the transcription factor RORγt. J Immunol. 2012;189(4):1765–1772. doi: 10.4049/jimmunol.1200858.
  • Carlson MJ, West ML, Coghill JM, Panoskaltsis-Mortari A, Blazar BR, Serody JS. In vitro-differentiated TH17 cells mediate lethal acute graft-versus-host disease with severe cutaneous and pulmonary pathologic manifestations. Blood. 2009;113(6):1365–1374. doi: 10.1182/blood-2008-06-162420.
  • Iclozan C, Yu Y, Liu C, Liang Y, Yi T, Anasetti C, Yu X-Z. T helper17 cells are sufficient but not necessary to induce acute graft-versus-host disease. Biol Blood Marrow Transplant. 2010;16(2):170–178. doi: 10.1016/j.bbmt.2009.09.023.
  • Mittrücker HW, Matsuyama T, Grossman A, Kündig TM, Potter J, Shahinian A, Wakeham A, Patterson B, Ohashi PS, Mak TW. Requirement for the transcription factor LSIRF/IRF4 for mature B and T lymphocyte function. Sci. 1997;275(5299):540–543. doi: 10.1126/science.275.5299.540.
  • Rengarajan J, Mowen KA, McBride KD, Smith ED, Singh H, Glimcher LH. Interferon regulatory factor 4 (IRF4) interacts with NFATc2 to modulate interleukin 4 gene expression. J Exp Med. 2002;195(8):1003–1012. doi: 10.1084/jem.20011128.
  • Honma K, Kimura D, Tominaga N, Miyakoda M, Matsuyama T, Yui K. Interferon regulatory factor 4 differentially regulates the production of Th2 cytokines in naive vs. effector/memory CD4+ T cells. Proc Natl Acad Sci U S A. 2008;105(41):15890–15895. doi: 10.1073/pnas.0803171105.
  • Tominaga N, Ohkusu-Tsukada K, Udono H, Abe R, Matsuyama T, Yui K. Development of Th1 and not Th2 immune responses in mice lacking IFN-regulatory factor-4. Int Immunol. 2003;15(1):1–10. doi: 10.1093/intimm/dxg001.
  • Lohoff M, Mittrücker H-W, Prechtl S, Bischof S, Sommer F, Kock S, Ferrick DA, Duncan GS, Gessner A, Mak TW. Dysregulated T helper cell differentiation in the absence of interferon regulatory factor 4. Proc Natl Acad Sci U S A. 2002;99(18):11808–11812. doi: 10.1073/pnas.182425099.
  • Staudt V, Bothur E, Klein M, Lingnau K, Reuter S, Grebe N, Gerlitzki B, Hoffmann M, Ulges A, Taube C, et al. 2010. Interferon-regulatory factor 4 is essential for the developmental program of T helper 9 cells. Immunity. 33(2):192–202. doi: 10.1016/j.immuni.2010.07.014.
  • Tamiya T, Ichiyama K, Kotani H, Fukaya T, Sekiya T, Shichita T, Honma K, Yui K, Matsuyama T, Nakao T, et al. Smad2/3 and IRF4 play a cooperative role in IL-9-producing T cell induction. J Immunol. 2013;191(5):2360–2371. doi: 10.4049/jimmunol.1301276.
  • Brüstle A, Heink S, Huber M, Rosenplänter C, Stadelmann C, Yu P, Arpaia E, Mak TW, Kamradt T, Lohoff M. The development of inflammatory T(H)-17 cells requires interferon-regulatory factor 4. Nat Immunol. 2007;8(9):958–966. doi: 10.1038/ni1500.
  • Huber M, Brüstle A, Reinhard K, Guralnik A, Walter G, Mahiny A, von LE, Lohoff M. IRF4 is essential for IL-21-mediated induction, amplification, and stabilization of the Th17 phenotype. Proc Natl Acad Sci U S A. 2008;105(52):20846–20851. doi: 10.1073/pnas.0809077106.
  • Kwon H, Thierry-Mieg D, Thierry-Mieg J, Kim H-P, Oh J, Tunyaplin C, Carotta S, Donovan CE, Goldman ML, Tailor P, et al. 2009. Analysis of interleukin-21-induced Prdm1 gene regulation reveals functional cooperation of STAT3 and IRF4 transcription factors. Immunity. 31(6):941–952. doi: 10.1016/j.immuni.2009.10.008.
  • Bollig N, Brüstle A, Kellner K, Ackermann W, Abass E, Raifer H, Camara B, Brendel C, Giel G, Bothur E, et al. Transcription factor IRF4 determines germinal center formation through follicular T-helper cell differentiation. Proc Natl Acad Sci U S A. 2012;109(22):8664–8669. doi: 10.1073/pnas.1205834109.
  • Mahnke J, Schumacher V, Ahrens S, Käding N, Feldhoff LM, Huber M, Rupp J, Raczkowski F, Mittrücker H-W. Interferon regulatory factor 4 controls TH1 cell effector function and metabolism. Sci Rep. 2016;6:35521. doi: 10.1038/srep35521.
  • Yang C, He D, Yin C, Tan J. Inhibition of interferon regulatory factor 4 suppresses Th1 and Th17 cell differentiation and ameliorates experimental autoimmune encephalomyelitis. Scand J Immunol. 2015;82(4):345–351. doi: 10.1111/sji.12334.
  • Wu J, Zhang H, Shi X, Xiao X, Fan Y, Minze LJ, Wang J, Ghobrial RM, Xia J, Sciammas R, et al. Ablation of transcription factor IRF4 promotes Transplant acceptance by driving allogenic CD4+ T cell dysfunction. Immunity. 2017;47(6):1114–1128.e6. doi: 10.1016/j.immuni.2017.11.003.
  • Vasanthakumar A, Moro K, Xin A, Liao Y, Gloury R, Kawamoto S, Fagarasan S, Mielke LA, Afshar-Sterle S, Masters SL, et al. The transcriptional regulators IRF4, BATF and IL-33 orchestrate development and maintenance of adipose tissue-resident regulatory T cells. Nat Immunol. 2015;16(3):276–285. doi: 10.1038/ni.3085.
  • Cretney E, Xin A, Shi W, Minnich M, Masson F, Miasari M, Belz GT, Smyth GK, Busslinger M, Nutt SL, et al. 2011. The transcription factors blimp-1 and IRF4 jointly control the differentiation and function of effector regulatory T cells. Nat Immunol. 12(4):304–311. doi: 10.1038/ni.2006.
  • Sidwell T, Liao Y, Garnham AL, Vasanthakumar A, Gloury R, Blume J, Teh PP, Chisanga D, Thelemann C, Rivera F de L, et al. Attenuation of TCR-induced transcription by Bach2 controls regulatory T cell differentiation and homeostasis. Nat Commun. 2020;11(1):252. doi: 10.1038/s41467-019-14112-2.
  • Cooke KR, Kobzik L, Martin TR, Brewer J, Delmonte J, Crawford JM, Ferrara JL. An experimental model of idiopathic pneumonia syndrome after bone marrow transplantation: I. The roles of minor H antigens and endotoxin. Blood. 1996;88(8):3230–3239. doi: 10.1182/blood.V88.8.3230.bloodjournal8883230.
  • Bolger AM, Lohse M, Usadel B. Trimmomatic: a flexible trimmer for Illumina sequence data. Bioinformatics. 2014;30(15):2114–2120. doi: 10.1093/bioinformatics/btu170.
  • Dobin A, Davis CA, Schlesinger F, Drenkow J, Zaleski C, Jha S, Batut P, Chaisson M, Gingeras TR. STAR: ultrafast universal RNA-seq aligner. Bioinformatics. 2013;29(1):15–21. doi: 10.1093/bioinformatics/bts635.
  • Li B, Dewey CN. RSEM: accurate transcript quantification from RNA-Seq data with or without a reference genome. BMC Bioinform. 2011;12:323. doi: 10.1186/1471-2105-12-323.
  • Love MI, Huber W, Anders S. Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2. Genome Biol. 2014;15(12):550. doi: 10.1186/s13059-014-0550-8.
  • Iwata A, Durai V, Tussiwand R, Briseño CG, Wu X, Grajales-Reyes GE, Egawa T, Murphy TL, Murphy KM. Quality of TCR signaling determined by differential affinities of enhancers for the composite BATF-IRF4 transcription factor complex. Nat Immunol. 2017;18(5):563–572. doi: 10.1038/ni.3714.
  • Lahmann A, Kuhrau J, Fuhrmann F, Heinrich F, Bauer L, Durek P, Mashreghi M-F, Hutloff A. Bach2 controls T follicular helper cells by direct repression of bcl-6. J Immunol. 2019;202(8):2229–2239. doi: 10.4049/jimmunol.1801400.
  • Huber M, Lohoff M. IRF4 at the crossroads of effector T-cell fate decision. Eur J Immunol. 2014;44(7):1886–1895. doi: 10.1002/eji.201344279.
  • Li P, Spolski R, Liao W, Wang L, Murphy TL, Murphy KM, Leonard WJ. BATF-JUN is critical for IRF4-mediated transcription in T cells. Nature. 2012;490(7421):543–546. doi: 10.1038/nature11530.
  • Mudter J, Amoussina L, Schenk M, Yu J, Brüstle A, Weigmann B, Atreya R, Wirtz S, Becker C, Hoffman A, et al. The transcription factor IFN regulatory factor-4 controls experimental colitis in mice via T cell-derived IL-6. J Clin Invest. 2008;118(7):2415–2426. doi: 10.1172/JCI33227.
  • Ullrich E, Abendroth B, Rothamer J, Huber C, Büttner-Herold M, Buchele V, Vogler T, Longerich T, Zundler S, Völkl S, et al. BATF-dependent IL-7RhiGM-CSF+ T cells control intestinal graft-versus-host disease. J Clin Invest. 2018;128(3):916–930. doi: 10.1172/JCI89242.
  • Raczkowski F, Ritter J, Heesch K, Schumacher V, Guralnik A, Höcker L, Raifer H, Klein M, Bopp T, Harb H, et al. The transcription factor Interferon regulatory factor 4 is required for the generation of protective effector CD8+ T cells. Proc Natl Acad Sci U S A. 2013;110(37):15019–15024. doi: 10.1073/pnas.1309378110.
  • Snook JP, Kim C, Williams MA. TCR signal strength controls the differentiation of CD4+ effector and memory T cells. Sci Immunol. 2018;3(25). doi: 10.1126/sciimmunol.aas9103.
  • Hojyo S, Tumes D, Murata A, Tokoyoda K. Multiple developmental pathways lead to the generation of CD4 T-cell memory. Int Immunol. 2020;32(9):589–595. doi: 10.1093/intimm/dxaa051.
  • Huang S, Shen Y, Pham D, Jiang L, Wang Z, Kaplan MH, Zhang G, Sun J. IRF4 modulates CD8+ T cell sensitivity to IL-2 family cytokines. Immunohorizons. 2017;1(6):92–100. doi: 10.4049/immunohorizons.1700020.
  • Zhang Y, Joe G, Hexner E, Zhu J, Emerson SG. Alloreactive memory T cells are responsible for the persistence of graft-versus-host disease. J Immunol. 2005;174(5):3051–3058. doi: 10.4049/jimmunol.174.5.3051.
  • Chennupati V, Held W. Feeling Exhausted? Tuning Irf4 Energizes Dysfunctional T Cells. Immunity. 2017;47(6):1009–1011. doi: 10.1016/j.immuni.2017.11.028.
  • Martinez RJ, Zhang N, Thomas SR, Nandiwada SL, Jenkins MK, Binstadt BA, Mueller DL. Arthritogenic self-reactive CD4+ T cells acquire an FR4hiCD73hi anergic state in the presence of Foxp3+ regulatory T cells. J Immunol. 2012;188(1):170–181. doi: 10.4049/jimmunol.1101311.
  • Crawford A, Angelosanto JM, Kao C, Doering TA, Odorizzi PM, Barnett BE, Wherry EJ. Molecular and transcriptional basis of CD4+ T cell dysfunction during chronic infection. Immunity. 2014;40(2):289–302. doi: 10.1016/j.immuni.2014.01.005.
  • Man K, Gabriel SS, Liao Y, Gloury R, Preston S, Henstridge DC, Pellegrini M, Zehn D, Berberich-Siebelt F, Febbraio MA, et al. Transcription factor IRF4 promotes CD8+ T cell exhaustion and limits the development of memory-like T cells during chronic infection. Immunity. 2017;47(6):1129–1141.e5. doi: 10.1016/j.immuni.2017.11.021.
  • Tkachev V, Kaminski J, Potter EL, Furlan SN, Yu A, Hunt DJ, McGuckin C, Zheng H, Colonna L, Gerdemann U, et al. Spatiotemporal single-cell profiling reveals that invasive and tissue-resident memory donor CD8+ T cells drive gastrointestinal acute graft-versus-host disease. Sci Transl Med. 2021;13(576). doi: 10.1126/scitranslmed.abc0227.
  • Roychoudhuri R, Hirahara K, Mousavi K, Clever D, Klebanoff CA, Bonelli M, Sciumè G, Zare H, Vahedi G, Dema B, et al. BACH2 represses effector programs to stabilize T(reg)-mediated immune homeostasis. Nature. 2013;498(7455):506–510. doi: 10.1038/nature12199.
  • Kim EH, Gasper DJ, Lee SH, Plisch EH, Svaren J, Suresh M. Bach2 regulates homeostasis of Foxp3+ regulatory T cells and protects against fatal lung disease in mice. J Immunol. 2014;192(3):985–995. doi: 10.4049/jimmunol.1302378.
  • Hu Q, Xu T, Zhang M, Zhang H, Liu Y, Li H-B, Chen C, Zheng J, Zhang Z, Li F, et al. Diverging regulation of Bach2 protein and RNA expression determine cell fate in early B cell response. Cell Rep. 2022;40(1):111035. doi: 10.1016/j.celrep.2022.111035.
  • Matsuyama T, Grossman A, Mittrücker HW, Siderovski DP, Kiefer F, Kawakami T, Richardson CD, Taniguchi T, Yoshinaga SK, Mak TW. Molecular cloning of LSIRF, a lymphoid-specific member of the interferon regulatory factor family that binds the interferon-stimulated response element (ISRE). Nucleic Acids Res. 1995;23(12):2127–2136. doi: 10.1093/nar/23.12.2127.
  • Itamura H, Shindo T, Tawara I, Kubota Y, Kariya R, Okada S, Komanduri KV, Kimura S. The MEK inhibitor trametinib separates murine graft-versus-host disease from graft-versus-tumor effects. JCI Insight. 2016;1(10):e86331. doi: 10.1172/jci.insight.86331.
  • Jankovic D, Ganesan J, Bscheider M, Stickel N, Weber FC, Guarda G, Follo M, Pfeifer D, Tardivel A, Ludigs K, et al. The Nlrp3 inflammasome regulates acute graft-versus-host disease. J Exp Med. 2013;210(10):1899–1910. doi: 10.1084/jem.20130084.