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:

A microRNA upregulated in asthma airway T cells promotes TH2 cytokine production

Abstract

MicroRNAs (miRNAs) exert powerful effects on immunological function by tuning networks of target genes that orchestrate cell activity. We sought to identify miRNAs and miRNA-regulated pathways that control the type 2 helper T cell (TH2 cell) responses that drive pathogenic inflammation in asthma. Profiling miRNA expression in human airway-infiltrating T cells revealed elevated expression of the miRNA miR-19a in asthma. Modulating miR-19 activity altered TH2 cytokine production in both human and mouse T cells, and TH2 cell responses were markedly impaired in cells lacking the entire miR-1792 cluster. miR-19 promoted TH2 cytokine production and amplified inflammatory signaling by direct targeting of the inositol phosphatase PTEN, the signaling inhibitor SOCS1 and the deubiquitinase A20. Thus, upregulation of miR-19a in asthma may be an indicator and a cause of increased TH2 cytokine production in the airways.

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: miR-19a expression is elevated in CD4+ T cells from asthmatic lungs.
Figure 2: The miR-1792 cluster promotes TH2 cytokine production.
Figure 3: The miR-1792 cluster promotes TH2 cytokine production in a cell-intrinsic and proliferation-independent manner.
Figure 4: miR-19a and miR-19b 'rescue' the TH2 cytokine defect in 1792Δ cells.
Figure 5: miR-19a promotes IL-13 production in human CD4+ T cells.
Figure 6: Several miR-19 targets negatively regulate TH2 cytokine production.
Figure 7: The miR-1792 cluster promotes TH2-driven inflammation in vivo.

Similar content being viewed by others

References

  1. Wenzel, S.E. Asthma phenotypes: the evolution from clinical to molecular approaches. Nat. Med. 18, 716–725 (2012).

    CAS  PubMed  Google Scholar 

  2. Locksley, R.M. Asthma and allergic inflammation. Cell 140, 777–783 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  3. Robinson, D.S. et al. Predominant TH2-like bronchoalveolar T-lymphocyte population in atopic asthma. N. Engl. J. Med. 326, 298–304 (1992).

    CAS  PubMed  Google Scholar 

  4. Woodruff, P.G. et al. T-helper Type 2–driven inflammation defines major subphenotypes of asthma. Am. J. Respir. Crit. Care Med. 180, 388–395 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  5. Wynn, T.A. IL-13 effector functions. Annu. Rev. Immunol. 21, 425–456 (2003).

    CAS  PubMed  Google Scholar 

  6. Kuperman, D.A. et al. Direct effects of interleukin-13 on epithelial cells cause airway hyperreactivity and mucus overproduction in asthma. Nat. Med. 8, 885–889 (2002).

    CAS  PubMed  Google Scholar 

  7. Corren, J. et al. Lebrikizumab treatment in adults with asthma. N. Engl. J. Med. 365, 1088–1098 (2011).

    CAS  PubMed  Google Scholar 

  8. Wenzel, S. et al. Dupilumab in persistent asthma with elevated eosinophil levels. N. Engl. J. Med. 368, 2455–2466 (2013).

    CAS  PubMed  Google Scholar 

  9. Ansel, K.M., Djuretic, I., Tanasa, B. & Rao, A. Regulation of Th2 differentiation and Il4 locus accessibility. Annu. Rev. Immunol. 24, 607–656 (2006).

    CAS  PubMed  Google Scholar 

  10. Bartel, D.P. MicroRNAs: target recognition and regulatory functions. Cell 136, 215–233 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  11. Liu, Y., Li, H., Xiao, T. & Lu, Q. Epigenetics in immune-mediated pulmonary diseases. Clin. Rev. Allergy Immunol. 45, 314–330 (2013).

    CAS  PubMed  Google Scholar 

  12. Solberg, O.D. et al. Airway epithelial miRNA expression is altered in asthma. Am. J. Respir. Crit. Care Med. 186, 965–974 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  13. Mattes, J., Collison, A., Plank, M., Phipps, S. & Foster, P.S. Antagonism of microRNA-126 suppresses the effector function of TH2 cells and the development of allergic airways disease. Proc. Natl. Acad. Sci. USA 106, 18704–18709 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  15. Baumjohann, D. & Ansel, K.M. MicroRNA-mediated regulation of T helper cell differentiation and plasticity. Nat. Rev. Immunol. 13, 666–678 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  16. Olive, V., Li, Q. & He, L. mir-17–92: a polycistronic oncomir with pleiotropic functions. Immunol. Rev. 253, 158–166 (2013).

    PubMed  PubMed Central  Google Scholar 

  17. Bronevetsky, Y. & Ansel, K.M. Regulation of miRNA biogenesis and turnover in the immune system. Immunol. Rev. 253, 304–316 (2013).

    PubMed  PubMed Central  Google Scholar 

  18. Jiang, S. et al. Molecular dissection of the miR-17–92 cluster's critical dual roles in promoting Th1 responses and preventing inducible Treg differentiation. Blood 118, 5487–5497 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  19. Xiao, C. et al. Lymphoproliferative disease and autoimmunity in mice with increased miR-17–92 expression in lymphocytes. Nat. Immunol. 9, 405–414 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  20. Steiner, D.F. et al. MicroRNA-29 regulates T-box transcription factors and interferon-γ production in helper T cells. Immunity 35, 169–181 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  21. Liu, S.-Q., Jiang, S., Li, C., Zhang, B. & Li, Q.-J. miR-17–92cluster targets phosphatase and tensin homology and Ikaros Family Zinc Finger 4 to promote TH17-mediated inflammation. J. Biol. Chem. 289, 12446–12456 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  22. de Kouchkovsky, D. et al. microRNA-17–92 regulates IL-10 production by regulatory T cells and control of experimental autoimmune encephalomyelitis. J. Immunol. 191, 1594–1605 (2013).

    CAS  PubMed  Google Scholar 

  23. Baumjohann, D. et al. The microRNA cluster miR-17–92 promotes TFH cell differentiation and represses subset-inappropriate gene expression. Nat. Immunol. 14, 840–848 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  24. Kang, S.G. et al. MicroRNAs of the miR-17–92 family are critical regulators of TFH differentiation. Nat. Immunol. 14, 849–857 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  25. Moltzahn, F. et al. Microfluidic-based multiplex qRT-PCR identifies diagnostic and prognostic microRNA signatures in the sera of prostate cancer patients. Cancer Res. 71, 550–560 (2011).

    CAS  PubMed  Google Scholar 

  26. Seumois, G. et al. An integrated nano-scale approach to profile miRNAs in limited clinical samples. Am. J. Clin. Exp. Immunol. 1, 70–89 (2012).

    PubMed  PubMed Central  Google Scholar 

  27. Bronevetsky, Y. et al. T cell activation induces proteasomal degradation of Argonaute and rapid remodeling of the microRNA repertoire. J. Exp. Med. 210, 417–432 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  28. Hsu, S.D. et al. miRTarBase: a database curates experimentally validated microRNA-target interactions. Nucleic Acids Res. 39, D163–D169 (2010).

    PubMed  PubMed Central  Google Scholar 

  29. Mavrakis, K.J. et al. Genome-wide RNA-mediated interference screen identifies miR-19 targets in Notch-induced T-cell acute lymphoblastic leukaemia. Nat. Cell Biol. 12, 372–379 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  30. Gantier, M.P. et al. A miR-19 regulon that controls NF-κB signaling. Nucleic Acids Res. 40, 8048–8058 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  31. Mingueneau, M. et al. The transcriptional landscape of αβ T cell differentiation. Nat. Immunol. 14, 619–632 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  32. Olive, V. et al. A component of the mir-17–92 polycistronic oncomir promotes oncogene-dependent apoptosis. eLife 2, e00822 (2013).

    PubMed  PubMed Central  Google Scholar 

  33. Srivastava, N., Sudan, R. & Kerr, W.G. Role of inositol poly-phosphatases and their targets in T cell biology. Front Immunol 4, 288 (2013).

    PubMed  PubMed Central  Google Scholar 

  34. Di Cristofano, A. et al. Impaired Fas response and autoimmunity in Pten+/− mice. Science 285, 2122–2125 (1999).

    CAS  PubMed  Google Scholar 

  35. Palmer, D.C. & Restifo, N.P. Suppressors of cytokine signaling (SOCS) in T cell differentiation, maturation, and function. Trends Immunol. 30, 592–602 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  36. Tanaka, K. et al. Loss of suppressor of cytokine signaling 1 in helper T cells leads to defective Th17 differentiation by enhancing antagonistic effects of IFN-gamma on STAT3 and Smads. J. Immunol. 180, 3746–3756 (2008).

    CAS  PubMed  Google Scholar 

  37. Egwuagu, C.E. et al. Suppressors of cytokine signaling proteins are differentially expressed in Th1 and Th2 cells: implications for Th cell lineage commitment and maintenance. J. Immunol. 168, 3181–3187 (2002).

    CAS  PubMed  Google Scholar 

  38. Fujimoto, M. et al. A regulatory role for suppressor of cytokine signaling-1 in Th polarization in vivo. Int. Immunol. 14, 1343–1350 (2002).

    CAS  PubMed  Google Scholar 

  39. Dickensheets, H. et al. Suppressor of cytokine signaling-1 is an IL-4-inducible gene in macrophages and feedback inhibits IL-4 signaling. Genes Immun. 8, 21–27 (2006).

    PubMed  Google Scholar 

  40. Tamiya, T., Kashiwagi, I., Takahashi, R., Yasukawa, H. & Yoshimura, A. Suppressors of cytokine signaling (SOCS) proteins and JAK/STAT pathways: regulation of T-cell inflammation by SOCS1 and SOCS3. Arterioscler. Thromb. Vasc. Biol. 31, 980–985 (2011).

    CAS  PubMed  Google Scholar 

  41. Coornaert, B. et al. T cell antigen receptor stimulation induces MALT1 paracaspase–mediated cleavage of the NF-κB inhibitor A20. Nat. Immunol. 9, 263–271 (2008).

    CAS  PubMed  Google Scholar 

  42. Liu, F., Qin, H.-B., Xu, B., Zhou, H. & Zhao, D.-Y. Profiling of miRNAs in pediatric asthma: upregulation of miRNA-221 and miRNA-485–3p. Mol Med Rep 6, 1178–1182 (2012).

    CAS  PubMed  Google Scholar 

  43. Bhakta, N.R. & Woodruff, P.G. Human asthma phenotypes: from the clinic, to cytokines, and back again. Immunol. Rev. 242, 220–232 (2011).

    PubMed  PubMed Central  Google Scholar 

  44. Rodriguez, A. et al. Requirement of bic/microRNA-155 for normal immune function. Science 316, 608–611 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  45. Thai, T.-H. et al. Regulation of the germinal center response by microRNA-155. Science 316, 604–608 (2007).

    CAS  PubMed  Google Scholar 

  46. Malmhäll, C. et al. MicroRNA-155 is essential for TH2-mediated allergen-induced eosinophilic inflammation in the lung. J. Allergy Clin. Immunol. 133, 1429–1438 e7 (2014).

    PubMed  Google Scholar 

  47. Collison, A., Mattes, J., Plank, M. & Foster, P.S. Inhibition of house dust mite-induced allergic airways disease by antagonism of microRNA-145 is comparable to glucocorticoid treatment. J. Allergy Clin. Immunol. 128, 160–167 e4 (2011).

    CAS  PubMed  Google Scholar 

  48. Kumar, M. et al. Let-7 microRNA-mediated regulation of IL-13 and allergic airway inflammation. J. Allergy Clin. Immunol. 128, 1077–85 e1–10 (2011).

    CAS  PubMed  Google Scholar 

  49. Sharma, A. et al. Antagonism of mmu-mir-106a attenuates asthma features in allergic murine model. J. Appl. Physiol. 113, 459–464 (2012).

    CAS  PubMed  Google Scholar 

  50. Edwards, M.R. et al. Targeting the NF-κB pathway in asthma and chronic obstructive pulmonary disease. Pharmacol. Ther. 121, 1–13 (2009).

    CAS  PubMed  Google Scholar 

  51. Suzuki, A. et al. T cell-specific loss of Pten leads to defects in central and peripheral tolerance. Immunity 14, 523–534 (2001).

    CAS  PubMed  Google Scholar 

  52. Chen, C. et al. Integrin α9β1 in airway smooth muscle suppresses exaggerated airway narrowing. J. Clin. Invest. 122, 2916–2927 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank M. McCune, Y. Bronevetsky and E. Krow-Lucal for help with human biospecimens, and R. Kageyama for discussion of the manuscript. Supported by the US National Institutes of Health (HL107202, HL109102), the Sandler Asthma Basic Research Center, a Scholar Award from The Leukemia & Lymphoma Society, a US National Science Foundation predoctoral fellowship (2010101500 to L.J.S.), the Swiss Foundation for Grants in Biology and Medicine (PASMP3-142725), the National Multiple Sclerosis Society and the UCSF Program for Breakthrough Biomedical Research, which is funded in part by the Sandler Foundation.

Author information

Authors and Affiliations

Authors

Contributions

L.J.S. designed, performed and analyzed most experiments. S.P., D.F.C., H.D.B., X.R., Y.W., H.H.P., D.B., M.M.M., M.P. and K.A.R. helped design and perform some experiments. X.H. helped design animal airway allergy experiments. N.R.B. analyzed human miRNA expression data. P.G.W., J.V.F. and J.R.A. designed and helped perform the clinical study for human miRNA expression analysis. K.M.A. helped design, analyze and interpret all experiments. L.J.S. and K.M.A. wrote the manuscript. All authors reviewed and approved the manuscript.

Corresponding author

Correspondence to K Mark Ansel.

Ethics declarations

Competing interests

D.F.C., H.D.B. and J.R.A. are employees of Genentech and are shareholders of the Roche Group. This work was supported in part by a grant to P.G.W. and J.V.F. from Genentech.

Integrated supplementary information

Supplementary Figure 1 Heatmap of all subjects and miRNAs analyzed by multiplex qPCR.

(a) Method shown for isolation of RNA from CD3+ CD4+ T cells from BAL fluid from healthy and asthmatic subjects. RNA was reverse transcribed and preamplified, and miRNA expression was analyzed using the Fluidigm Biomark qPCR system. (b) Unsupervised hierarchical clustering using the 23 most variable miRNAs (between healthy and steroid-naïve asthma) was determined in R (version 2.14.1) using the hclust function with “complete” method. The heatmap (R version 2.14.1; heatmap function) was generated using the hierarchical clustering above, including expression of all miRNAs determined by multiplex qPCR using the Biomark system (Fluidigm). In the heatmap, red indicates expression level higher than the mean across all subjects, blue denotes expression level lower than the mean, and grey indicates expression not determined (N.D). In the first row, green indicates asthmatic subjects, and yellow indicates healthy subjects. In the second row, purple indicates inhaled corticosteroid (ICS) treatment, and white indicates no ICS. miRNAs are arranged by average expression across all subjects (lowest to highest). Significance was determined by Welch t-test comparing the healthy and SN asthma groups. (Full table of p-values is provided in Supplementary Table 2.)

Supplementary Figure 2 miR-1792 expression in 1792Δ and 1792+ naive CD4 T cells.

(a) Schematic of the genomic loci of the miR-1792, miR-106a363, and miR-106b25 clusters. Colors indicate miRNAs belonging to the same family; yellow = miR-17 family, blue = miR-18 family, teal = miR-19 family, pink = miR-92 family. (b) miRNA expression was determined by SYBR qPCR in 1792+/+ (black bars) and 1792Δ/Δ (white bars) naïve CD4 T cells. Expression was normalized to 5.8S rRNA in each sample. Data are representative of 2 experiments.

Supplementary Figure 3 miR-19 activity in 1792Δ and 1792+ cells and effects of mimics and inhibitors.

(a) DGCR8+/+ and DGCR8Δ/Δ CD4 T cells transduced with retroviral sensors expressing GFP with 4 perfectly complementary binding sites for miR-1 (psens1), miR-19a (psens19a), or miR-19b (psens19b) in the 3’UTR. (b) DGCR8Δ/Δ CD4 T cells transduced with miR-19a (psens19a) or miR-19b (psens19b) sensors and transfected with control mimic (CM), miR-19a mimic (19a), or miR-19b mimic (19b). (c) DGCR8+/+ CD4 T cells transduced with psens19a or psens19b, and transfected with control inhibitor (CI) or anti-miR-19a and anti-miR-19b inhibitors (anti-19ab). (d) Analysis of cytokine expression by flow cytometry in DGCR8+/+ CD4 T cells cultured for 5 days in nonpolarizing conditions and transfected with control inhibitor (CI), and anti-miR-19b inhibitors (anti-19). Data represent mean +/- SEM. * p = 0.03. (e) DGCR8+/+ CD4 T cells cultured for 5 days in nonpolarizing conditions and transfected with control mimic (CM), miR-19a mimic (19a), or miR-19b mimic (19b). Data represent mean +/- SEM.

Supplementary Figure 4 Effects of miR-1792 members on proliferation.

Analysis (day 5) of cell proliferation by flow cytometry of Th2-polarized cells labeled with CellTrace Violet (CTV). Grey filled histogram represents 1792+/+ cells transfected with control mimic (CM), black histogram represents 1792Δ/Δ cells transfected with control mimic (CM), and red histogram represents cells transfected with the corresponding miRNA mimic. Data are representative of 3 independent experiments, with 3 technical replicates in each experiment.

Supplementary Figure 5 Identification of myeloid cells in BAL fluid in an in vivo allergic airway inflammation model.

(a) Method for TH2 OT-II cell adoptive transfer model of allergic airway inflammation. See Materials and Methods for details. (b) Gating strategy used to identify eosinophils (CD45+ CD11b+ Siglec F+), neutrophils (CD45+ CD11b+ Ly6G+), and alveolar macrophages (CD45+ CD11c+ Siglec F+) in bronchoalveolar lavage (BAL) fluid from mice that received 1792+/+ or 1792 Δ/Δ OT-II Th2 cells and were challenged with ovalbumin for 3 consecutive days. Cells are first gated as CD45+, Live (eF780 viability dye-/CD4-/CD8-/CD19-), singlet, NK1.1-, and then divided into the above mentioned cell types. Numbers of each cell type are determined by the product of the frequency of that cell type (as a percentage of live cells) and the total BAL cell count determined by Coulter Counter.

Supplementary information

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Simpson, L., Patel, S., Bhakta, N. et al. A microRNA upregulated in asthma airway T cells promotes TH2 cytokine production. Nat Immunol 15, 1162–1170 (2014). https://doi.org/10.1038/ni.3026

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/ni.3026

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