1,472
Views
8
CrossRef citations to date
0
Altmetric
Review

Regulation of mRNA stability by RBPs and noncoding RNAs contributing to the pathogenicity of Th17 cells

, ORCID Icon, , , , , , & show all
Pages 647-656 | Received 14 Aug 2020, Accepted 07 Dec 2020, Published online: 23 Dec 2020

References

  • Matsuzaki G, Umemura M. Interleukin-17 as an effector molecule of innate and acquired immunity against infections. Microbiol Immunol. 2007;51(12):1139–1147.
  • Kolls JK, Khader SA. The role of Th17 cytokines in primary mucosal immunity. Cytokine Growth Factor Rev. 2010;21:443–448.
  • Dong C. TH17 cells in development: an updated view of their molecular identity and genetic programming. Nat Rev Immunol. 2008;8:337–348.
  • Harrington LE, Hatton RD, Mangan PR, et al. Interleukin 17-producing CD4+ effector T cells develop via a lineage distinct from the T helper type 1 and 2 lineages. Nat Immunol. 2005;6:1123–1132.
  • Korn T, Bettelli E, Oukka M, et al. IL-17 and Th17 Cells. Annu Rev Immunol. 2009;27:485–517.
  • Veldhoen M, Hocking RJ, Atkins CJ, et al. TGFbeta in the context of an inflammatory cytokine milieu supports de novo differentiation of IL-17-producing T cells. Immunity. 2006;24:179–189.
  • Bettelli E, Carrier Y, Gao W, et al. Reciprocal developmental pathways for the generation of pathogenic effector TH17 and regulatory T cells. Nature. 2006;441:235–238.
  • Mangan PR, Harrington LE, O’Quinn DB, et al. Transforming growth factor-beta induces development of the T(H)17 lineage. Nature. 2006;441:231–234.
  • Yosef N, Shalek AK, Gaublomme JT, et al. Dynamic regulatory network controlling TH17 cell differentiation. Nature. 2013;496:461–468.
  • Zhou L, Chong MMW, Littman DR. Plasticity of CD4+ T cell lineage differentiation. Immunity. 2009;30(5):646–655.
  • Zhu J, Yamane H, Paul WE. Differentiation of effector CD4 T cell populations (*). Annu Rev Immunol. 2010;28:445–489.
  • Yasuda K, Takeuchi Y, Hirota K. The pathogenicity of Th17 cells in autoimmune diseases. Semin Immunopathol. 2019;41:283–297.
  • Wilke CM, Bishop K, Fox D, et al. Deciphering the role of Th17 cells in human disease. Trends Immunol. 2011;32:603–611.
  • Kafasla P, Skliris A, Kontoyiannis DL. Post-transcriptional coordination of immunological responses by RNA-binding proteins. Nat Immunol. 2014;15:492–502.
  • Bjur E, Larsson O, Yurchenko E, et al. Distinct translational control in CD4+ T cell subsets. PLoS Genet. 2013;9:e1003494.
  • Ciofani M, Madar A, Galan C, et al. A validated regulatory network for Th17 cell specification. Cell. 2012;151:289–303.
  • Bhatt DM, Pandya-Jones A, Tong AJ, et al. Transcript dynamics of proinflammatory genes revealed by sequence analysis of subcellular RNA fractions. Cell. 2012;150:279–290.
  • Hao S, Baltimore D. The stability of mRNA influences the temporal order of the induction of genes encoding inflammatory molecules. Nat Immunol. 2009;10:281–288.
  • Hao S, Baltimore D. RNA splicing regulates the temporal order of TNF-induced gene expression. Proc Natl Acad Sci U S A. 2013;110:11934–11939.
  • Garneau NL, Wilusz J, Wilusz CJ. The highways and byways of mRNA decay. Nat Rev Mol Cell Biol. 2007;8:113–126.
  • Le Hir H, Izaurralde E, Maquat LE, et al. The spliceosome deposits multiple proteins 20-24 nucleotides upstream of mRNA exon-exon junctions. Embo J. 2000;19:6860–6869.
  • Diaz-Munoz MD, Turner M. Uncovering the role of RNA-Binding proteins in gene expression in the immune system. Front Immunol. 2018;9:1094.
  • Matsushita K, Takeuchi O, Standley DM, et al. Zc3h12a is an RNase essential for controlling immune responses by regulating mRNA decay. Nature. 2009;458:1185–1190.
  • Yang C, Huang S, Wang X, et al. Emerging roles of CCCH-Type zinc finger proteins in destabilizing mRNA encoding inflammatory factors and regulating immune responses. Crit Rev Eukaryot Gene Expr. 2015;25:77–89.
  • Uehata T, Iwasaki H, Vandenbon A, et al. Malt1-induced cleavage of regnase-1 in CD4(+) helper T cells regulates immune activation. Cell. 2013;153:1036–1049.
  • Minagawa K, Wakahashi K, Kawano H, et al. Posttranscriptional modulation of cytokine production in T cells for the regulation of excessive inflammation by TFL. J Iimmunol. 2014;192:1512–1524.
  • Yoshinaga M, Takeuchi O. RNA binding proteins in the control of autoimmune diseases. Immunological medicine. 2019;42:53–64.
  • Athanasopoulos V, Ramiscal RR, Vinuesa CG. ROQUIN signalling pathways in innate and adaptive immunity. Eur J Immunol. 2016;46:1082–1090.
  • Lin AE, Mak TW. The role of E3 ligases in autoimmunity and the regulation of autoreactive T cells. Curr Opin Immunol. 2007;19:665–673.
  • Leppek K, Schott J, Reitter S, et al. Roquin promotes constitutive mRNA decay via a conserved class of stem-loop recognition motifs. Cell. 2013;153:869–881.
  • Fu M, Blackshear PJ. RNA-binding proteins in immune regulation: a focus on CCCH zinc finger proteins. Nat Rev Immunol. 2017;17:130–143.
  • Sgromo A, Raisch T, Bawankar P, et al. A CAF40-binding motif facilitates recruitment of the CCR4-NOT complex to mRNAs targeted by Drosophila Roquin. Nat Commun. 2017;8:14307.
  • Yu H, Pardoll D, Jove R. STATs in cancer inflammation and immunity: a leading role for STAT3. Nat Rev Cancer. 2009;9:798–809.
  • Flanagan SE, Haapaniemi E, Russell MA, et al. Activating germline mutations in STAT3 cause early-onset multi-organ autoimmune disease. Nat Genet. 2014;46:812–814.
  • Fogli LK, Sundrud MS, Goel S, et al. T cell-derived IL-17 mediates epithelial changes in the airway and drives pulmonary neutrophilia. J Iimmunol. 2013;191:3100–3111.
  • Masuda K, Ripley B, Nyati KK, et al. Arid5a regulates naive CD4+ T cell fate through selective stabilization of Stat3 mRNA. J Exp Med. 2016;213:605–619.
  • Jeltsch KM, Hu D, Brenner S, et al. Cleavage of roquin and regnase-1 by the paracaspase MALT1 releases their cooperatively repressed targets to promote T(H)17 differentiation. Nat Immunol. 2014;15:1079–1089.
  • Watts TH. TNF/TNFR family members in costimulation of T cell responses. Annu Rev Immunol. 2005;23:23–68.
  • Xiao X, Shi X, Fan Y, et al. The costimulatory receptor OX40 inhibits Interleukin-17 expression through activation of repressive chromatin remodeling pathways. Immunity. 2016;44:1271–1283.
  • Demirci G, Amanullah F, Kewalaramani R, et al. Critical role of OX40 in CD28 and CD154-independent rejection. J Iimmunol. 2004;172:1691–1698.
  • Piconese S, Valzasina B, Colombo MP. OX40 triggering blocks suppression by regulatory T cells and facilitates tumor rejection. J Exp Med. 2008;205:825–839.
  • Xiao X, Balasubramanian S, Liu W, et al. OX40 signaling favors the induction of T(H)9 cells and airway inflammation. Nat Immunol. 2012;13:981–990.
  • Fu Y, Lin Q, Zhang Z, et al. Therapeutic strategies for the costimulatory molecule OX40 in T-cell-mediated immunity. Acta Pharm Sin B. 2020;10:414–433.
  • Hanieh H, Masuda K, Metwally H, et al. Arid5a stabilizes OX40 mRNA in murine CD4(+) T cells by recognizing a stem-loop structure in its 3ʹUTR. Eur J Immunol. 2018;48:593–604.
  • Yamazaki S, Muta T, Takeshige K. A novel IkappaB protein, IkappaB-zeta, induced by proinflammatory stimuli, negatively regulates nuclear factor-kappaB in the nuclei. J Biol Chem. 2001;276:27657–27662.
  • Okamoto K, Iwai Y, Oh-Hora M, et al. IkappaBzeta regulates T(H)17 development by cooperating with ROR nuclear receptors. Nature. 2010;464:1381–1385.
  • Muta T. IkappaB-zeta: an inducible regulator of nuclear factor-kappaB. Vitam Horm. 2006;74:301–316.
  • Essig K, Kronbeck N, Guimaraes JC, et al. Roquin targets mRNAs in a 3ʹ-UTR-specific manner by different modes of regulation. Nat Commun. 2018;9:3810.
  • Wawro M, Wawro K, Kochan J, et al. ZC3H12B/MCPIP2, a new active member of the ZC3H12 family. Rna. 2019;25:840–856.
  • Paulos CM, Carpenito C, Plesa G, et al. The inducible costimulator (ICOS) is critical for the development of human T(H)17 cells. Sci Transl Med. 2010;2:55ra78.
  • Rudd CE, Schneider H. Unifying concepts in CD28, ICOS and CTLA4 co-receptor signalling. Nat Rev Immunol. 2003;3:544–556.
  • Brunet JF, Denizot F, Luciani MF, et al. A new member of the immunoglobulin superfamily–CTLA-4. Nature. 1987;328:267–270.
  • Chikuma S, Bluestone JA. CTLA-4 and tolerance: the biochemical point of view. Immunol Res. 2003;28:241–253.
  • Tivol EA, Borriello F, Schweitzer AN, et al. Loss of CTLA-4 leads to massive lymphoproliferation and fatal multiorgan tissue destruction, revealing a critical negative regulatory role of CTLA-4. Immunity. 1995;3:541–547.
  • Ying H, Yang L, Qiao G, et al. Cutting edge: CTLA-4–B7 interaction suppresses Th17 cell differentiation. J Immunol. 2010;185:1375–1378.
  • Hinman MN, Lou H. Diverse molecular functions of Hu proteins. Cell Mol Life Sci. 2008;65:3168–3181.
  • Pabis M, Popowicz GM, Stehle R, et al. HuR biological function involves RRM3-mediated dimerization and RNA binding by all three RRMs. Nucleic Acids Res. 2019;47:1011–1029.
  • Fan XC, Steitz JA. Overexpression of HuR, a nuclear-cytoplasmic shuttling protein, increases the in vivo stability of ARE-containing mRNAs. Embo J. 1998;17:3448–3460.
  • Peng SS, Chen CY, Xu N, et al. RNA stabilization by the AU-rich element binding protein, HuR, an ELAV protein. Embo J. 1998;17:3461–3470.
  • Ma WJ, Chung S, Furneaux H. The Elav-like proteins bind to AU-rich elements and to the poly(A) tail of mRNA. Nucleic Acids Res. 1997;25:3564–3569.
  • Chen J, Martindale JL, Cramer C, et al. The RNA-binding protein HuR contributes to neuroinflammation by promoting C-C chemokine receptor 6 (CCR6) expression on Th17 cells. J Biol Chem. 2017;292:14532–14543.
  • Chen J, Cascio J, Magee JD, et al. Posttranscriptional gene regulation of IL-17 by the RNA-binding protein HuR is required for initiation of experimental autoimmune encephalomyelitis. J Iimmunol. 2013;191:5441–5450.
  • Maity A, McKenna WG, Muschel RJ. Cyclin A message stability varies with the cell cycle. Cell growth differentiation. 1997;8:311–318.
  • Howe JA, Howell M, Hunt T, et al. Identification of a developmental timer regulating the stability of embryonic cyclin A and a new somatic A-type cyclin at gastrulation. Genes Dev. 1995;9:1164–1176.
  • Trembley JH, Kren BT, Steer CJ. Posttranscriptional regulation of cyclin B messenger RNA expression in the regenerating rat liver. Cell growth differentiation. 1994;5:99–108.
  • Wang W, Caldwell MC, Lin S, et al. HuR regulates cyclin A and cyclin B1 mRNA stability during cell proliferation. EMBO J. 2000;19:2340–2350.
  • El-Behi M, Ciric B, Dai H, et al. The encephalitogenicity of T(H)17 cells is dependent on IL-1- and IL-23-induced production of the cytokine GM-CSF. Nat Immunol. 2011;12:568–575.
  • Codarri L, Gyulveszi G, Tosevski V, et al. RORgammat drives production of the cytokine GM-CSF in helper T cells, which is essential for the effector phase of autoimmune neuroinflammation. Nat Immunol. 2011;12:560–567.
  • Carrieri PB, Provitera V, De Rosa T, et al. Profile of cerebrospinal fluid and serum cytokines in patients with relapsing-remitting multiple sclerosis: a correlation with clinical activity. Immunopharmacol Immunotoxicol. 1998;20:373–382.
  • Perrella O, Carrieri PB, De Mercato R, et al. Markers of activated T lymphocytes and T cell receptor gamma/delta+ in patients with multiple sclerosis. Eur Neurol. 1993;33:152–155.
  • Shiomi A, Usui T. Pivotal roles of GM-CSF in autoimmunity and inflammation. Mediators Inflamm. 2015;2015:568543.
  • Constantinescu CS, Asher A, Fryze W, et al. Randomized phase 1b trial of MOR103, a human antibody to GM-CSF, in multiple sclerosis. Neurol Neuroimmunol Neuroinflamm. 2015;2:e117.
  • Chen J, Adamiak W, Huang G, et al. Interaction of RNA-binding protein HuR and miR-466i regulates GM-CSF expression. Sci Rep. 2017;7:17233.
  • Maddur MS, Miossec P, Kaveri SV, et al. Th17 cells: biology, pathogenesis of autoimmune and inflammatory diseases, and therapeutic strategies. Am J Pathol. 2012;181:8–18.
  • Zaballos A, Varona R, Gutierrez J, et al. Molecular cloning and RNA expression of two new human chemokine receptor-like genes. Biochem Biophys Res Commun. 1996;227:846–853.
  • Schutyser E, Struyf S, Van Damme J. The CC chemokine CCL20 and its receptor CCR6. Cytokine Growth Factor Rev. 2003;14:409–426.
  • Reboldi A, Coisne C, Baumjohann D, et al. C-C chemokine receptor 6-regulated entry of TH-17 cells into the CNS through the choroid plexus is required for the initiation of EAE. Nat Immunol. 2009;10:514–523.
  • Blackshear PJ. Tristetraprolin and other CCCH tandem zinc-finger proteins in the regulation of mRNA turnover. Biochem Soc Trans. 2002;30:945–952.
  • Lai WS, Carballo E, Thorn JM, et al. Interactions of CCCH zinc finger proteins with mRNA. Binding of tristetraprolin-related zinc finger proteins to Au-rich elements and destabilization of mRNA. J Biol Chem. 2000;275:17827–17837.
  • Lai WS, Stumpo DJ, Wells ML, et al. Importance of the conserved carboxyl-terminal CNOT1 binding domain to tristetraprolin activity in vivo. Mol Cell Biol. 2019;39.
  • Lee HH, Yoon NA, Vo MT, et al. Tristetraprolin down-regulates IL-17 through mRNA destabilization. FEBS Lett. 2012;586:41–46.
  • Lykke-Andersen J, Wagner E. Recruitment and activation of mRNA decay enzymes by two ARE-mediated decay activation domains in the proteins TTP and BRF-1. Genes Dev. 2005;19:351–361.
  • Fenger-Gron M, Fillman C, Norrild B, et al. Multiple processing body factors and the ARE binding protein TTP activate mRNA decapping. Mol Cell. 2005;20:905–915.
  • Peng H, Ning H, Wang Q, et al. Tristetraprolin regulates TH17 cell function and ameliorates DSS-induced colitis in mice. Front Immunol. 2020;11:1952.
  • Xiao C, Rajewsky K. MicroRNA control in the immune system: basic principles. Cell. 2009;136:26–36.
  • Jonas S, Izaurralde E. Towards a molecular understanding of microRNA-mediated gene silencing. Nat Rev Genet. 2015;16:421–433.
  • Iwakawa HO, Tomari Y. The functions of MicroRNAs: mRNA decay and translational repression. Trends Cell Biol. 2015;25:651–665.
  • Arribas-Hernandez L, Kielpinski LJ, Brodersen P. mRNA decay of most arabidopsis miRNA targets requires slicer activity of AGO1. Plant Physiol. 2016;171:2620–2632.
  • Fukaya T, Tomari Y. MicroRNAs mediate gene silencing via multiple different pathways in drosophila. Mol Cell. 2012;48:825–836.
  • Narozna B, Langwinski W, Szczepankiewicz A. Non-Coding RNAs in pediatric airway diseases. Genes (Basel). 2017;8.
  • Zhu YJ, Mao D, Gao W, et al. Peripheral whole blood lncRNA expression analysis in patients with eosinophilic asthma. Medicine (Baltimore). 2018;97:e9817.
  • Qiu YY, Wu Y, Lin MJ, et al. LncRNA-MEG3 functions as a competing endogenous RNA to regulate Treg/Th17 balance in patients with asthma by targeting microRNA-17/RORgammat. Biomed Pharmacothe. 2019;111:386–394.
  • Russell-Puleri S, Dela Paz NG, Adams D, et al. Fluid shear stress induces upregulation of COX-2 and PGI2 release in endothelial cells via a pathway involving PECAM-1, PI3K, FAK, and p38. Am J Physiol Heart Circ Physiol. 2017;312:H485–H500.
  • Li H, Bradbury JA, Dackor RT, et al. Cyclooxygenase-2 regulates Th17 cell differentiation during allergic lung inflammation. Am J Respir Crit Care Med. 2011;184:37–49.
  • Qiu L, Zhang Y, Do DC, et al. miR-155 modulates cockroach allergen- and oxidative stress-induced cyclooxygenase-2 in asthma. J Iimmunol. 2018;201:916–929.
  • Hedrick MN, Lonsdorf AS, Shirakawa AK, et al. CCR6 is required for IL-23-induced psoriasis-like inflammation in mice. J Clin Invest. 2009;119:2317–2329.
  • Hirota K, Yoshitomi H, Hashimoto M, et al. Preferential recruitment of CCR6-expressing Th17 cells to inflamed joints via CCL20 in rheumatoid arthritis and its animal model. J Exp Med. 2007;204:2803–2812.
  • Yamazaki T, Yang XO, Chung Y, et al. CCR6 regulates the migration of inflammatory and regulatory T cells. J Iimmunol. 2008;181:8391–8401.
  • Murugaiyan G, da Cunha AP, Ajay AK, et al. MicroRNA-21 promotes Th17 differentiation and mediates experimental autoimmune encephalomyelitis. J Clin Invest. 2015;125:1069–1080.
  • Ross SH, Cantrell DA. Signaling and function of Interleukin-2 in T lymphocytes. Annu Rev Immunol. 2018;36(1):411–433.
  • Li Q, Zhang D, Wang Y, et al. MiR-21/Smad 7 signaling determines TGF-beta1-induced CAF formation. Sci Rep. 2013;3:2038.
  • Yang L, Boldin MP, Yu Y, et al. miR-146a controls the resolution of T cell responses in mice. J Exp Med. 2012;209:1655–1670.
  • Oeckinghaus A, Hayden MS, Ghosh S. Crosstalk in NF-kappaB signaling pathways. Nat Immunol. 2011;12:695–708.
  • Li B, Wang X, Choi IY, et al. miR-146a modulates autoreactive Th17 cell differentiation and regulates organ-specific autoimmunity. J Clin Invest. 2017;127:3702–3716.
  • Suddason T, Gallagher E. Genetic insights into Map3k-dependent proliferative expansion of T cells. Cell Cycle. 2016;15:1956–1960.
  • Chen L, Al-Mossawi MH, Ridley A, et al. miR-10b-5p is a novel Th17 regulator present in Th17 cells from ankylosing spondylitis. Ann Rheum Dis. 2017;76:620–625.
  • Wei GH, Wang X. lncRNA MEG3 inhibit proliferation and metastasis of gastric cancer via p53 signaling pathway. Eur Rev Med Pharmacol Sci. 2017;21:3850–3856.
  • Uroda T, Anastasakou E, Rossi A, et al. Conserved pseudoknots in lncRNA MEG3 are essential for stimulation of the p53 pathway. Mol Cell. 2019;75:982–95 e9.
  • Wu MX, Ao Z, Prasad KV, et al. IEX-1L, an apoptosis inhibitor involved in NF-kappaB-mediated cell survival. Science. 1998;281:998–1001.
  • Zhang Y, Schlossman SF, Edwards RA, et al. Impaired apoptosis, extended duration of immune responses, and a lupus-like autoimmune disease in IEX-1-transgenic mice. Proc Natl Acad Sci U S A. 2002;99:878–883.
  • Zhi L, Ustyugova IV, Chen X, et al. Enhanced Th17 differentiation and aggravated arthritis in IEX-1-deficient mice by mitochondrial reactive oxygen species-mediated signaling. J Iimmunol. 2012;189:1639–1647.
  • Liu Z, Liu L, Zhong Y, et al. LncRNA H19 over-expression inhibited Th17 cell differentiation to relieve endometriosis through miR-342-3p/IER3 pathway. Cell Biosci. 2019;9:84.
  • Qi M, Zhou Q, Zeng W, et al. Analysis of long non-coding RNA expression of lymphatic endothelial cells in response to type 2 diabetes. Cell Physiol Biochem. 2017;41:466–474.
  • Hou J, Wang L, Wu Q, et al. Long noncoding RNA H19 upregulates vascular endothelial growth factor A to enhance mesenchymal stem cells survival and angiogenic capacity by inhibiting miR-199a-5p. Stem Cell Res Ther. 2018;9:109.
  • Wang SH, Ma F, Tang ZH, et al. Long non-coding RNA H19 regulates FOXM1 expression by competitively binding endogenous miR-342-3p in gallbladder cancer. J Exp Clin Cancer Res. 2016;35:160.
  • Ji L, Zhan Y, Hua F, et al. The ratio of Treg/Th17 cells correlates with the disease activity of primary immune thrombocytopenia. PloS One. 2012;7:e50909.
  • Lee HT, Shiao YM, Wu TH, et al. Serum BLC/CXCL13 concentrations and renal expression of CXCL13/CXCR5 in patients with systemic lupus erythematosus and lupus nephritis. J Rheumatol. 2010;37:45–52.
  • Burkle A, Niedermeier M, Schmitt-Graff A, et al. Overexpression of the CXCR5 chemokine receptor, and its ligand, CXCL13 in B-cell chronic lymphocytic leukemia. Blood. 2007;110:3316–3325.
  • Jernas M, Nookaew I, Wadenvik H, et al. MicroRNA regulate immunological pathways in T-cells in immune thrombocytopenia (ITP). Blood. 2013;121:2095–2098.
  • Li JQ, Hu SY, Wang ZY, et al. MicroRNA-125-5p targeted CXCL13: a potential biomarker associated with immune thrombocytopenia. Am J Transl Res. 2015;7:772–780.
  • Li JQ, Hu SY, Wang ZY, et al. Long non-coding RNA MEG3 inhibits microRNA-125a-5p expression and induces immune imbalance of Treg/Th17 in immune thrombocytopenic purpura. Biomed Pharmacothe. 2016;83:905–911.
  • Li HB, Tong J, Zhu S, et al. m(6)A mRNA methylation controls T cell homeostasis by targeting the IL-7/STAT5/SOCS pathways. Nature. 2017;548:338–342.
  • Yang XP, Ghoreschi K, Steward-Tharp SM, et al. Opposing regulation of the locus encoding IL-17 through direct, reciprocal actions of STAT3 and STAT5. Nat Immunol. 2011;12:247–254.
  • Wang CX, Cui GS, Liu X, et al. METTL3-mediated m6A modification is required for cerebellar development. PLoS Biol. 2018;16:e2004880.
  • Meisel M, Hermann-Kleiter N, Hinterleitner R, et al. The kinase PKCalpha selectively upregulates interleukin-17A during Th17 cell immune responses. Immunity. 2013;38:41–52.
  • Eberhardt W, Badawi A, Biyanee A, et al. Cytoskeleton-dependent transport as a potential target for interfering with post-transcriptional HuR mRNA regulons. Front Pharmacol. 2016;7:251.
  • Liang J, Wang J, Azfer A, et al. A novel CCCH-zinc finger protein family regulates proinflammatory activation of macrophages. J Biol Chem. 2008;283:6337–6346.
  • Ansa-Addo EA, Huang HC, Riesenberg B, et al. RNA binding protein PCBP1 is an intracellular immune checkpoint for shaping T cell responses in cancer immunity. Sci Adv. 2020;6:eaaz3865.
  • Ren J, Li B. The functional stability of FOXP3 and RORgammat in Treg and Th17 and their therapeutic applications. Adv Protein Chem Struct Biol. 2017;107:155–189.

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.