Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Article
  • Published:

The microRNA miR-22 inhibits the histone deacetylase HDAC4 to promote TH17 cell–dependent emphysema

Abstract

Smoking-related emphysema is a chronic inflammatory disease driven by the TH17 subset of helper T cells through molecular mechanisms that remain obscure. Here we explored the role of the microRNA miR-22 in emphysema. We found that miR-22 was upregulated in lung myeloid dendritic cells (mDCs) of smokers with emphysema and antigen-presenting cells (APCs) of mice exposed to smoke or nanoparticulate carbon black (nCB) through a mechanism that involved the transcription factor NF-κB. Mice deficient in miR-22, but not wild-type mice, showed attenuated TH17 responses and failed to develop emphysema after exposure to smoke or nCB. We further found that miR-22 controlled the activation of APCs and TH17 responses through the activation of AP-1 transcription factor complexes and the histone deacetylase HDAC4. Thus, miR-22 is a critical regulator of both emphysema and TH17 responses.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Figure 1: Enhanced miR-22 expression in human emphysema and cigarette smoke–activated lung APCs.
Figure 2: miR-22 deficiency protects mice from nCB-induced emphysema.
Figure 3: miR-22 expression is transcriptionally regulated through NF-κB and AP-1 complexes.
Figure 4: miR-22 promotes the activation of lung APCs.
Figure 5: miR-22 promotes the transcriptional activity of AP-1 in APCs.
Figure 6: miR-22 targets HDAC4 in APCs to promote IL-6 production.
Figure 7: HDAC4 inhibits the activation of APCs and the progression of emphysema.
Figure 8: Neutralization of lung miR-22 reverses nCB-induced emphysema.

Similar content being viewed by others

Accession codes

Primary accessions

Gene Expression Omnibus

References

  1. Hoyert, D.L. & Xu, J.Q. Deaths: preliminary data for 2011. Natl. Vital Stat. Rep. 61, 51–65 (2012).

    Google Scholar 

  2. Wilson, D.O. et al. Association of radiographic emphysema and airflow obstruction with lung cancer. Am. J. Respir. Crit. Care Med. 178, 738–744 (2008).

    Article  PubMed  PubMed Central  Google Scholar 

  3. Hart, J.E., Eisen, E.A. & Laden, F. Occupational diesel exhaust exposure as a risk factor for chronic obstructive pulmonary disease. Curr. Opin. Pulm. Med. 18, 151–154 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Xu, C. et al. Autoreactive T cells in human smokers is predictive of clinical outcome. Front. Immunol. 3, 267 (2012).

    Article  PubMed  PubMed Central  Google Scholar 

  5. Grumelli, S. et al. An immune basis for lung parenchymal destruction in chronic obstructive pulmonary disease and emphysema. PLoS Med. 1, 75–83 (2004).

    Article  CAS  Google Scholar 

  6. Shan, M. et al. Lung myeloid dendritic cells coordinately induce TH1 and TH17 responses in human emphysema. Sci. Transl. Med. 1, 28 (2009).

    Article  CAS  Google Scholar 

  7. Shan, M. et al. Agonistic induction of PPAR-γ reverses cigarette smoke induced emphysema. J. Clin. Invest. 124, 1371–1381 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Shan, M. et al. Cigarette smoke induction of osteopontin (SPP1) mediates TH17 inflammation in human and experimental emphysema. Sci. Transl. Med. 4, 18 (2012).

    Article  CAS  Google Scholar 

  9. O'Connell, R.M., Rao, D.S., Chaudhuri, A.A. & Baltimore, D. Physiological and pathological roles for microRNAs in the immune system. Nat. Rev. Immunol. 10, 111–122 (2010).

    Article  CAS  PubMed  Google Scholar 

  10. Polikepahad, S. et al. Proinflammatory role for let-7 microRNAS in experimental asthma. J. Biol. Chem. 285, 30139–30149 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. You, R. et al. Nanoparticulate carbon black in cigarette smoke induce DNA cleavage and Th17-mediated emphysema. eLife (in the press).

  12. Rincon, M. & Irvin, C.G. Role of IL-6 in asthma and other inflammatory pulmonary diseases. Int. J. Biol. Sci. 8, 1281–1290 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Rodriguez, A., Griffiths-Jones, S., Ashurst, J.L. & Bradley, A. Identification of mammalian microRNA host genes and transcription units. Genome Res. 14, 1902–1910 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Hou, Y., Moreau, F. & Chadee, K. PPARγ is an E3 ligase that induces the degradation of NF-κB/p65. Nat. Commun. 3, 1300 (2012).

    Article  CAS  PubMed  Google Scholar 

  15. Shan, M. et al. Agonistic induction of PPARγ reverses cigarette smoke-induced emphysema. J. Clin. Invest. 124, 1371–1381 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Greenlee, K.J., Werb, Z. & Kheradmand, F. Matrix metalloproteinases in lung: multiple, multifarious, and multifaceted. Physiol. Rev. 87, 69–98 (2007).

    Article  CAS  PubMed  Google Scholar 

  17. O'Sullivan, B.J. & Thomas, R. CD40 ligation conditions dendritic cell antigen-presenting function through sustained activation of NF-kB. J. Immunol. 168, 5491–5498 (2002).

    Article  CAS  PubMed  Google Scholar 

  18. Mann, J., Oakley, F., Johnson, P.W. & Mann, D.A. CD40 induces interleukin-6 gene transcription in dendritic cells: regulation by TRAF2, AP-1, NF-κB, AND CBF1. J. Biol. Chem. 277, 17125–17138 (2002).

    Article  CAS  PubMed  Google Scholar 

  19. Oeckinghaus, A., Hayden, M.S. & Ghosh, S. Crosstalk in NF-κB signaling pathways. Nat. Immunol. 12, 695–708 (2011).

    Article  CAS  PubMed  Google Scholar 

  20. Huang, Z.P. et al. MicroRNA-22 regulates cardiac hypertrophy and remodeling in response to stress. Circ. Res. 112, 1234–1243 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Jovicic, A., Zaldivar Jolissaint, J.F., Moser, R., Silva Santos Mde, F. & Luthi-Carter, R. MicroRNA-22 (miR-22) overexpression is neuroprotective via general anti-apoptotic effects and may also target specific Huntington's disease-related mechanisms. PLoS ONE 8, e54222 (2013).

  22. Zhang, J. et al. microRNA-22, downregulated in hepatocellular carcinoma and correlated with prognosis, suppresses cell proliferation and tumourigenicity. Br. J. Cancer 103, 1215–1220 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. de Ruijter, A.J., van Gennip, A.H., Caron, H.N., Kemp, S. & van Kuilenburg, A.B. Histone deacetylases (HDACs): characterization of the classical HDAC family. Biochem. J. 370, 737–749 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Parra, M. Class IIa HDACs - new insights into their functions in physiology and pathology. FEBS J. 282, 1736–1744 (2015).

    Article  CAS  PubMed  Google Scholar 

  25. Luan, B. et al. Leptin-mediated increases in catecholamine signaling reduce adipose tissue inflammation via activation of macrophage HDAC4. Cell Metab. 19, 1058–1065 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Gordon, J.W. et al. Protein kinase A-regulated assembly of a MEF2-HDAC4 repressor complex controls c-Jun expression in vascular smooth muscle cells. J. Biol. Chem. 284, 19027–19042 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Wang, A.H. et al. HDAC4, a human histone deacetylase related to yeast HDA1, is a transcriptional corepressor. Mol. Cell. Biol. 19, 7816–7827 (1999).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Gurha, P. et al. Targeted deletion of microRNA-22 promotes stress-induced cardiac dilation and contractile dysfunction. Circulation 125, 2751–2761 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Gurha, P. et al. microRNA-22 promotes heart failure through coordinate suppression of PPAR/ERR-nuclear hormone receptor transcription. PLoS ONE 8, e75882 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Huang, S. et al. Upregulation of miR-22 promotes osteogenic differentiation and inhibits adipogenic differentiation of human adipose tissue-derived mesenchymal stem cells by repressing HDAC6 protein expression. Stem Cells Dev. 21, 2531–2540 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Tang, Y. et al. microRNA-22 acts as a metastasis suppressor by targeting metadherin in gastric cancer. Mol. Med. Rep. 11, 454–460 (2015).

    Article  CAS  PubMed  Google Scholar 

  32. Xu, Q.F. et al. MiR-22 is frequently downregulated in medulloblastomas and inhibits cell proliferation via the novel target PAPST1. Brain Pathol. 24, 568–583 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Song, S.J. et al. The oncogenic microRNA miR-22 targets the TET2 tumor suppressor to promote hematopoietic stem cell self-renewal and transformation. Cell Stem Cell 13, 87–101 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Keatings, V.M., Jatakanon, A., Worsdell, Y.M. & Barnes, P.J. Effects of inhaled and oral glucocorticoids on inflammatory indices in asthma and COPD. Am. J. Respir. Crit. Care Med. 155, 542–548 (1997).

    Article  CAS  PubMed  Google Scholar 

  35. Mizuno, S. et al. Inhibition of histone deacetylase causes emphysema. Am. J. Physiol. Lung Cell. Mol. Physiol. 300, L402–L413 (2011).

    Article  CAS  PubMed  Google Scholar 

  36. Lemercier, C. et al. Class II histone deacetylases are directly recruited by BCL6 transcriptional repressor. J. Biol. Chem. 277, 22045–22052 (2002).

    Article  CAS  PubMed  Google Scholar 

  37. Sandhu, S.K. et al. miR-155 targets histone deacetylase 4 (HDAC4) and impairs transcriptional activity of B-cell lymphoma 6 (BCL6) in the Emu-miR-155 transgenic mouse model. Proc. Natl. Acad. Sci. USA 109, 20047–20052 (2012).

    Article  PubMed  PubMed Central  Google Scholar 

  38. Han, S. et al. Recruitment of histone deacetylase 4 by transcription factors represses interleukin-5 transcription. Biochem. J. 400, 439–448 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Yang, Q. et al. Cross talk between histone deacetylase 4 and STAT6 in the transcriptional regulation of arginase 1 during mouse dendritic cell differentiation. Mol. Cell. Biol. 35, 63–75 (2015).

    Article  CAS  PubMed  Google Scholar 

  40. Liu, W. et al. AP-1 activated by toll-like receptors regulates expression of IL-23 p19. J. Biol. Chem. 284, 24006–24016 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Wang, X. et al. Early secreted antigenic target of 6-kDa protein of Mycobacterium tuberculosis primes dendritic cells to stimulate Th17 and inhibit Th1 immune responses. J. Immunol. 189, 3092–3103 (2012).

    Article  CAS  PubMed  Google Scholar 

  42. Bettelli, E. et al. Reciprocal developmental pathways for the generation of pathogenic effector TH17 and regulatory T cells. Nature 441, 235–238 (2006).

    CAS  PubMed  Google Scholar 

  43. Ivanov, I.I. et al. The orphan nuclear receptor RORγt directs the differentiation program of proinflammatory IL-17+ T helper cells. Cell 126, 1121–1133 (2006).

    Article  CAS  PubMed  Google Scholar 

  44. Kudo, M. et al. IL-17A produced by αβ T cells drives airway hyper-responsiveness in mice and enhances mouse and human airway smooth muscle contraction. Nat. Med. 18, 547–554 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Röhn, T.A. et al. Vaccination against IL-17 suppresses autoimmune arthritis and encephalomyelitis. Eur. J. Immunol. 36, 2857–2867 (2006).

    Article  CAS  PubMed  Google Scholar 

  46. Elson, C.O. et al. Monoclonal anti-interleukin 23 reverses active colitis in a T cell-mediated model in mice. Gastroenterology 132, 2359–2370 (2007).

    Article  CAS  PubMed  Google Scholar 

  47. Hirota, K. et al. T cell self-reactivity forms a cytokine milieu for spontaneous development of IL-17+ Th cells that cause autoimmune arthritis. J. Exp. Med. 204, 41–47 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Majdzadeh, N. et al. HDAC4 inhibits cell-cycle progression and protects neurons from cell death. Dev. Neurobiol. 68, 1076–1092 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Pauwels, R.A. et al. Global strategy for the diagnosis, management, and prevention of chronic obstructive pulmonary disease. NHLBI/WHO Global Initiative for Chronic Obstructive Lung Disease (GOLD) Workshop summary. Am. J. Respir. Crit. Care Med. 163, 1256–1276 (2001).

    Article  CAS  PubMed  Google Scholar 

  50. Corry, D.B. et al. Interleukin 4, but not interleukin 5 or eosinophils, is required in a murine model of acute airway hyperreactivity. J. Exp. Med. 183, 109–117 (1996).

    Article  CAS  PubMed  Google Scholar 

  51. Lee, S.H. et al. Differential requirement for CD18 in T-helper effector homing. Nat. Med. 9, 1281–1286 (2003).

    Article  CAS  PubMed  Google Scholar 

  52. Corry, D.B. et al. Decreased allergic lung inflammatory cell egression and increased susceptibility to asphyxiation in MMP2-deficiency. Nat. Immunol. 3, 347–353 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank J. Levitt (Baylor College of Medicine) for CD11c-Cre mice; E. Olson (UT Southwestern Medical Center) for Hdac4fl mice; and W. Decker, J. Sederstrom, Y. Qian and L.-Z. Song for technical assistance. Supported by the US National Institutes of Health (R01HL117181 to F.K.; R01HL110883 to F.K. and D.B.C.; and AI036211, CA125123 and RR024574 to the Cytometry and Cell Sorting Core at Baylor College of Medicine) and the US Veterans Administration Office of Research and Development (1I01BX002221 to D.B.C.).

Author information

Authors and Affiliations

Authors

Contributions

W.L. and D.B.C. conceptualized the project and all studies and wrote the manuscript with assistance from all co-authors; W.L. designed and performed all experiments, with R.Y., X.Y. and T.Y. contributing to selected mouse experiments and micro-CT analysis; E.L.G.S., D.C.M., W.K.A.S. and J.M.T. prepared nCB; A.R. provided miR-22 deficient mice; and F.K. and D.B.C. provided grant support.

Corresponding authors

Correspondence to Farrah Kheradmand or David B Corry.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Integrated supplementary information

Supplementary Figure 1 miR-22 is required for airway MMP activities but not for the development of γδ+ T cells in emphysema.

(a) Gelatin zymogram depicting matrix metalloproteinase (MMP) 2 and MMP9 activities in bronchoalveolar lavage fluid (BALF) of wild type (WT; lanes 3-5) and Mir22–/– mice (lanes 6-9) previously challenged with nCB. Lanes 1 and 2 are BALF from naïve wild type mice. (b) Total lung IL-17A+ γδ+ T cells were quantified by flow cytometry.

Supplementary Figure 2 Mir22–/– mice develop reduced TH17 responses upon sensitization with ovalbumin.

Both WT and Mir22–/– mice were intraperitoneally injected with 25µg ovalbumin precipitated in alum weekly for three consecutive weeks. Total splenocytes were harvested at the end of the fifth week. (a) IFN-γ, (b) IL-4 and (c) IL-17A positive cells with ovalbumin (OVA; 0.5 mg/ml overnight) and without (Media) were quantified by ELISpot. *: p<0.05, Kruskal Wallis test. n=4.

Supplementary Figure 3 nCB-induced production of pro-inflammatory cytokines and chemokines from whole lung is in part miR-22 dependent.

WT and Mir22–/– mice were challenged intranasally with nCB over one month. (a-d) Lungs were then collected and the concentrations of the indicated cytokines and chemokines from lung homogenate fluids were determined by Bio-plex. (e-g) Lung CD11c+ cells were isolated and cultured ex vivo overnight. Concentrations of the indicated supernatant chemokines were determined by Bio-plex. *: p<0.05; **: p<0.01, ***: p<0.001, Kruskal Wallis test. n=4-5.

Supplementary Figure 4 Smoke-exposed wild-type lung APCs are sufficient to induce emphysema in mice with and without miR-22.

Lung CD11c+ APC from four month cigarette SMK or air exposed WT mice were isolated and adaptively transferred to WT and Mir22–/– recipients. (a) Micro-CT quantification of recipient mouse lung volume after 3 months. (b) Total macrophages in BALF. (c) Relative abundance of lung IL-17A+ TH17 cells as assessed by flow cytometry. *: p<0.05; **: p<0.01, Kruskal Wallis test. n=3-4 as indicated in (A).

Supplementary Figure 5 PPAR-γ regulates miR-22 expression in lung APCs.

(a) MiR-22 expression in CD11c+ lung APCs from eight month old Ppargflox and PpargCD11c mice. (b) MiR-22 expression in CD11c+ lung APCs from air or cigarette smoke (SMK) exposed mice with or without intranasally challenge of ciglitazone (Cig). *: p<0.05; unpaired t-test in (a) one-way ANOVA test in (b). (n=3)

Supplementary Figure 6 miR-22 is required for pro-inflammatory gene expression in nCB-exposed lung APCs.

Lung CD11c+ cells from WT and Mir22–/– mice exposed to PBS or nCB were isolated. Expression of indicated genes was determined by quantitative PCR. *, p<0.05; **, p<0.01; ***, p<0.001, One-way ANOVA test. (n=3)

Supplementary Figure 7 miR-22 deficient APCs induce a TH17 cytokine in vitro with supplementation of IL-6.

MiR-22 sufficient (Mir22flox) and deficient BMDCs (Mir22CD11c) were primed with or without 1000ng nCB and co-cultured with WT naïve CD4+ T cells for 3 days and secreted IL-17A was measured by ELISA (n=4-5). 10ng/ml IL-6 supplement was added as indicated. *: p<0.05; Mann-Whitney test (n=4-5).

Supplementary Figure 8 Silencing miR-22 alters pro- and anti-inflammatory gene expression in APCs.

Mice were sacrificed at the 5 month point in Figure (8a). (a) MiR-22 expression level in lung CD11c+ cells. IL-6 (b) and IL-17A (c) concentration in lung homogenates as assessed by Bioplex. (d-f) Indicated gene expression in isolated lung CD11c+ cells determined by RT-qPCR. *, p<0.05; **, p<0.01; ***, p<0.001, Kruskal Wallis test (n=4-6). Data in are from one of two comparable experiments.

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–8 (PDF 1105 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Lu, W., You, R., Yuan, X. et al. The microRNA miR-22 inhibits the histone deacetylase HDAC4 to promote TH17 cell–dependent emphysema. Nat Immunol 16, 1185–1194 (2015). https://doi.org/10.1038/ni.3292

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/ni.3292

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing