DOI QR코드

DOI QR Code

Vitamin C enhances the expression of IL17 in a Jmjd2-dependent manner

  • Song, Mi Hye (Department of Pathology, Hallym University College of Medicine) ;
  • Nair, Varun Sasidharan (Department of Pathology, Hallym University College of Medicine) ;
  • Oh, Kwon Ik (Department of Pathology, Hallym University College of Medicine)
  • Received : 2016.11.21
  • Accepted : 2016.12.08
  • Published : 2017.01.31

Abstract

Previously, we reported that vitamin C facilitates the CpG demethylation of Foxp3 enhancer in $CD4^+Foxp3^+$ regulatory T cells (Tregs) by enhancing the activity of a DNA demethylase ten-eleven-translocation (Tet). However, it is not clear whether vitamin C affects other helper T cell lineages like T helper type 17 (Th17) cells which are related with Tregs. Here, we show that the expression of interleukin-17A (IL17) increases with the treatment of vitamin C but not with other antioxidants. Interestingly, the upregulation of IL17 was not accompanied by DNA demethylation in Il17 promoter and was independent of Tet enzymes. Rather, vitamin C reduced the trimethylation of histone H3 lysine 9 (H3K9me3) in the regulatory elements of the Il17 locus, and the effects of vitamin C were abrogated by knockdown of jumonji-C domain-containing protein 2 (jmjd2). These results suggest that vitamin C can affect the expression of IL17 by modulating the histone demethylase activity.

Keywords

References

  1. Korn T, Bettelli E, Oukka M and Kuchroo VK (2009) IL-17 and Th17 Cells. Annu Rev Immunol 27, 485-517 https://doi.org/10.1146/annurev.immunol.021908.132710
  2. Mangan PR, Harrington LE, O'Quinn DB et al (2006) Transforming growth factor-beta induces development of the T(H)17 lineage. Nature 441, 231-234 https://doi.org/10.1038/nature04754
  3. Ciofani M, Madar A, Galan C et al (2012) A validated regulatory network for Th17 cell specification. Cell 151, 289-303 https://doi.org/10.1016/j.cell.2012.09.016
  4. Durant L, Watford WT, Ramos HL et al (2010) Diverse targets of the transcription factor STAT3 contribute to T cell pathogenicity and homeostasis. Immunity 32, 605-615 https://doi.org/10.1016/j.immuni.2010.05.003
  5. Ivanov II, McKenzie BS, Zhou L et al (2006) The orphan nuclear receptor RORgammat directs the differentiation program of proinflammatory IL-17+ T helper cells. Cell 126, 1121-1133 https://doi.org/10.1016/j.cell.2006.07.035
  6. Patel DD and Kuchroo VK (2015) Th17 Cell Pathway in Human Immunity: Lessons from Genetics and Therapeutic Interventions. Immunity 43, 1040-1051. https://doi.org/10.1016/j.immuni.2015.12.003
  7. Young JI, Zuchner S and Wang G (2015) Regulation of the Epigenome by Vitamin C. Annu Rev Nutr 35, 545-564 https://doi.org/10.1146/annurev-nutr-071714-034228
  8. Gorres KL and Raines RT (2010) Prolyl 4-hydroxylase. Crit Rev Biochem Mol Biol 45, 106-124 https://doi.org/10.3109/10409231003627991
  9. Sasidharan Nair V, Song MH and Oh KI (2016) Vitamin C Facilitates Demethylation of the Foxp3 Enhancer in a Tet-Dependent Manner. J Immunol 196, 2119-2131 https://doi.org/10.4049/jimmunol.1502352
  10. Yue X, Trifari S, Aijo T et al (2016) Control of Foxp3 stability through modulation of TET activity. J Exp Med 213, 377-397 https://doi.org/10.1084/jem.20151438
  11. Dickson KM, Gustafson CB, Young JI, Zuchner S and Wang G (2013) Ascorbate-induced generation of 5-hydroxymethylcytosine is unaffected by varying levels of iron and 2-oxoglutarate. Biochem Biophys Res Commun 439, 522-527 https://doi.org/10.1016/j.bbrc.2013.09.010
  12. Xu GL and Walsh CP (2014) Enzymatic DNA oxidation: mechanisms and biological significance. BMB Rep 47, 609-618 https://doi.org/10.5483/BMBRep.2014.47.11.223
  13. Chen J, Liu H, Liu J et al (2013) H3K9 methylation is a barrier during somatic cell reprogramming into iPSCs. Nat Genet 45, 34-42 https://doi.org/10.1038/ng.2491
  14. Knowles HJ, Raval RR, Harris AL and Ratcliffe PJ (2003) Effect of ascorbate on the activity of hypoxia-inducible factor in cancer cells. Cancer Res 63, 1764-1768
  15. Baek SH and Kim KI (2016) Regulation of HIF-1alpha stability by lysine methylation. BMB Rep 49, 245-246 https://doi.org/10.5483/BMBRep.2016.49.5.053
  16. Hopkinson RJ, Tumber A, Yapp C et al (2013) 5-Carboxy-8-hydroxyquinoline is a Broad Spectrum 2-Oxoglutarate Oxygenase Inhibitor which Causes Iron Translocation. Chem Sci 4, 3110-3117 https://doi.org/10.1039/c3sc51122g
  17. Schofield CJ and Ratcliffe PJ (2004) Oxygen sensing by HIF hydroxylases. Nat Rev Mol Cell Biol 5, 343-354
  18. Chowdhury R, Yeoh KK, Tian YM et al (2011) The oncometabolite 2-hydroxyglutarate inhibits histone lysine demethylases. EMBO Rep 12, 463-469 https://doi.org/10.1038/embor.2011.43
  19. Dang EV, Barbi J, Yang HY et al (2011) Control of T(H)17/T(reg) balance by hypoxia-inducible factor 1. Cell 146, 772-784 https://doi.org/10.1016/j.cell.2011.07.033
  20. Yang BH, Floess S, Hagemann S et al (2015) Development of a unique epigenetic signature during in vivo Th17 differentiation. Nucleic Acids Res 43, 1537-1548 https://doi.org/10.1093/nar/gkv014
  21. Ichiyama K, Chen T, Wang X et al (2015) The methylcytosine dioxygenase Tet2 promotes DNA demethylation and activation of cytokine gene expression in T cells. Immunity 42, 613-626 https://doi.org/10.1016/j.immuni.2015.03.005
  22. Blaschke K, Ebata KT, Karimi MM et al (2013) Vitamin C induces Tet-dependent DNA demethylation and a blastocyst-like state in ES cells. Nature 500, 222-226 https://doi.org/10.1038/nature12362
  23. Li Q, Zou J, Wang M et al (2014) Critical role of histone demethylase Jmjd3 in the regulation of CD4+ T-cell differentiation. Nat Commun 5, 5780 https://doi.org/10.1038/ncomms6780
  24. Liu Z, Cao W, Xu L et al (2015) The histone H3 lysine-27 demethylase Jmjd3 plays a critical role in specific regulation of Th17 cell differentiation. J Mol Cell Biol 7, 505-516 https://doi.org/10.1093/jmcb/mjv022
  25. Xiao X, Shi X, Fan Y et al (2016) The Costimulatory Receptor OX40 Inhibits Interleukin-17 Expression through Activation of Repressive Chromatin Remodeling Pathways. Immunity 44, 1271-1283 https://doi.org/10.1016/j.immuni.2016.05.013
  26. Hojfeldt JW, Agger K and Helin K (2013) Histone lysine demethylases as targets for anticancer therapy. Nat Rev Drug Discov 12, 917-930 https://doi.org/10.1038/nrd4154
  27. Pedersen MT, Kooistra SM, Radzisheuskaya A et al (2016) Continual removal of H3K9 promoter methylation by Jmjd2 demethylases is vital for ESC self-renewal and early development. EMBO J 35, 1550-1564 https://doi.org/10.15252/embj.201593317
  28. Seong MA, Woo JK, Kang JH et al (2015) Oral administration of fermented wild ginseng ameliorates DSS-induced acute colitis by inhibiting NF-kappaB signaling and protects intestinal epithelial barrier. BMB Rep 48, 419-425 https://doi.org/10.5483/BMBRep.2015.48.7.039
  29. Mantei A, Rutz S, Janke M et al (2008) siRNA stabilization prolongs gene knockdown in primary T lymphocytes. Eur J Immunol 38, 2616-2625 https://doi.org/10.1002/eji.200738075
  30. Zuo J, Ma H, Cai H, Wu Y, Jiang W and Yu L (2015) An inhibitory role of NEK6 in TGFbeta/Smad signaling pathway. BMB Rep 48, 473-478 https://doi.org/10.5483/BMBRep.2015.48.8.225

Cited by

  1. Modulating epigenetic memory through vitamins and TET: implications for regenerative medicine and cancer treatment vol.9, pp.6, 2017, https://doi.org/10.2217/epi-2017-0021
  2. Alloantigen-Induced Regulatory T Cells Generated in Presence of Vitamin C Display Enhanced Stability of Foxp3 Expression and Promote Skin Allograft Acceptance vol.8, 2017, https://doi.org/10.3389/fimmu.2017.00748
  3. Stochastics of Cellular Differentiation Explained by Epigenetics: The Case of T-Cell Differentiation and Functional Plasticity 2017, https://doi.org/10.1111/sji.12589
  4. Influence of Vitamin C on Lymphocytes: An Overview vol.7, pp.3, 2018, https://doi.org/10.3390/antiox7030041
  5. Vitamin C and immune cell function in inflammation and cancer vol.46, pp.5, 2018, https://doi.org/10.1042/BST20180169
  6. Intravenous Vitamin C for Cancer Therapy – Identifying the Current Gaps in Our Knowledge vol.9, pp.1664-042X, 2018, https://doi.org/10.3389/fphys.2018.01182
  7. Vitamin C Fosters the In Vivo Differentiation of Peripheral CD4+ Foxp3− T Cells into CD4+ Foxp3+ Regulatory T Cells but Impairs Their Ability to Prolong Skin Allograft Survival vol.9, pp.1664-3224, 2018, https://doi.org/10.3389/fimmu.2018.00112