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:

Drosha controls dendritic cell development by cleaving messenger RNAs encoding inhibitors of myelopoiesis

Abstract

To investigate if the microRNA (miRNA) pathway is required for dendritic cell (DC) development, we assessed the effect of ablating Drosha and Dicer, the two enzymes central to miRNA biogenesis. We found that while Dicer deficiency had some effect, Drosha deficiency completely halted DC development and halted myelopoiesis more generally. This indicated that while the miRNA pathway did have a role, it was a non-miRNA function of Drosha that was particularly critical. Drosha repressed the expression of two mRNAs encoding inhibitors of myelopoiesis in early hematopoietic progenitors. We found that Drosha directly cleaved stem-loop structure within these mRNAs and that this mRNA degradation was necessary for myelopoiesis. We have therefore identified a mechanism that regulates the development of DCs and other myeloid cells.

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: Drosha deficiency perturbs myeloid populations more severely than does Dicer deficiency.
Figure 2: Drosha deficiency impairs the DC development of cells cultured with Flt3L, but Dicer deficiency does not.
Figure 3: Drosha deficiency impairs DC development at an early stage of myelopoiesis.
Figure 4: The requirement for Drosha in myelopoiesis is not circumvented by inflammatory or cytokine challenge.
Figure 5: Gene-expression profiling of Drosha- or Dicer-deficient LSK cells.
Figure 6: Derepression of Myl9 or Todr1 in HSCs is responsible for the DC-developmental block in Drosha deficient mice.
Figure 7: The inhibitors of myelopoiesis Myl9 and Todr1 are direct endonucleolytic targets of Drosha in hematopoietic progenitors.

Similar content being viewed by others

Accession codes

Primary accessions

Gene Expression Omnibus

References

  1. Kondo, M., Weissman, I.L. & Akashi, K. Identification of clonogenic common lymphoid progenitors in mouse bone marrow. Cell 91, 661–672 (1997).

    Article  CAS  PubMed  Google Scholar 

  2. Akashi, K., Traver, D., Miyamoto, T. & Weissman, I.L. A clonogenic common myeloid progenitor that gives rise to all myeloid lineages. Nature 404, 193–197 (2000).

    Article  CAS  PubMed  Google Scholar 

  3. Naik, S.H. et al. Development of plasmacytoid and conventional dendritic cell subtypes from single precursor cells derived in vitro and in vivo. Nat. Immunol. 8, 1217–1226 (2007).

    Article  CAS  PubMed  Google Scholar 

  4. Onai, N. et al. Identification of clonogenic common Flt3+M-CSFR+ plasmacytoid and conventional dendritic cell progenitors in mouse bone marrow. Nat. Immunol. 8, 1207–1216 (2007).

    Article  CAS  PubMed  Google Scholar 

  5. Corcoran, L. et al. The lymphoid past of mouse plasmacytoid cells and thymic dendritic cells. J. Immunol. 170, 4926–4932 (2003).

    Article  CAS  PubMed  Google Scholar 

  6. D'Amico, A. & Wu, L. The early progenitors of mouse dendritic cells and plasmacytoid predendritic cells are within the bone marrow hemopoietic precursors expressing Flt3. J. Exp. Med. 198, 293–303 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Onai, N., Obata-Onai, A., Tussiwand, R., Lanzavecchia, A. & Manz, M.G. Activation of the Flt3 signal transduction cascade rescues and enhances type I interferon-producing and dendritic cell development. J. Exp. Med. 203, 227–238 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Carotta, S. et al. The transcription factor PU.1 controls dendritic cell development and Flt3 cytokine receptor expression in a dose-dependent manner. Immunity 32, 628–641 (2010).

    Article  CAS  PubMed  Google Scholar 

  9. Belz, G.T. & Nutt, S.L. Transcriptional programming of the dendritic cell network. Nat. Rev. Immunol. 12, 101–113 (2012).

    Article  CAS  PubMed  Google Scholar 

  10. Chong, M.M. et al. Canonical and alternate functions of the microRNA biogenesis machinery. Genes Dev. 24, 1951–1960 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Koralov, S.B. et al. Dicer ablation affects antibody diversity and cell survival in the B lymphocyte lineage. Cell 132, 860–874 (2008).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Lee, Y. et al. The nuclear RNase III Drosha initiates microRNA processing. Nature 425, 415–419 (2003).

    Article  CAS  PubMed  Google Scholar 

  14. Grishok, A. et al. Genes and mechanisms related to RNA interference regulate expression of the small temporal RNAs that control C. elegans developmental timing. Cell 106, 23–34 (2001).

    Article  CAS  PubMed  Google Scholar 

  15. Hutvágner, G. et al. A cellular function for the RNA-interference enzyme Dicer in the maturation of the let-7 small temporal RNA. Science 293, 834–838 (2001).

    Article  PubMed  Google Scholar 

  16. Ketting, R.F. et al. Dicer functions in RNA interference and in synthesis of small RNA involved in developmental timing in C. elegans. Genes Dev. 15, 2654–2659 (2001).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Li, H.S., Greeley, N., Sugimoto, N., Liu, Y.J. & Watowich, S.S. miR-22 controls Irf8 mRNA abundance and murine dendritic cell development. PLoS ONE 7, e52341 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Mildner, A. et al. Mononuclear phagocyte miRNome analysis identifies miR-142 as critical regulator of murine dendritic cell homeostasis. Blood 121, 1016–1027 (2013).

    Article  CAS  PubMed  Google Scholar 

  19. Alemdehy, M.F. et al. Dicer1 deletion in myeloid-committed progenitors causes neutrophil dysplasia and blocks macrophage/dendritic cell development in mice. Blood 119, 4723–4730 (2012).

    Article  CAS  PubMed  Google Scholar 

  20. Guo, S. et al. MicroRNA miR-125a controls hematopoietic stem cell number. Proc. Natl. Acad. Sci. USA 107, 14229–14234 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Kuipers, H., Schnorfeil, F.M. & Brocker, T. Differentially expressed microRNAs regulate plasmacytoid vs. conventional dendritic cell development. Mol. Immunol. 48, 333–340 (2010).

    Article  CAS  PubMed  Google Scholar 

  22. Gantier, M.P. et al. Analysis of microRNA turnover in mammalian cells following Dicer1 ablation. Nucleic Acids Res. 39, 5692–5703 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Merad, M. & Manz, M.G. Dendritic cell homeostasis. Blood 113, 3418–3427 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Brasel, K., De Smedt, T., Smith, J.L. & Maliszewski, C.R. Generation of murine dendritic cells from flt3-ligand-supplemented bone marrow cultures. Blood 96, 3029–3039 (2000).

    Article  CAS  PubMed  Google Scholar 

  25. Han, J. et al. Posttranscriptional crossregulation between Drosha and DGCR8. Cell 136, 75–84 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Karginov, F.V. et al. Diverse endonucleolytic cleavage sites in the mammalian transcriptome depend upon microRNAs, Drosha, and additional nucleases. Mol. Cell 38, 781–788 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Kirigin, F.F. et al. Dynamic microRNA gene transcription and processing during T cell development. J. Immunol. 188, 3257–3267 (2012).

    Article  CAS  PubMed  Google Scholar 

  28. Zuker, M. Mfold web server for nucleic acid folding and hybridization prediction. Nucleic Acids Res. 31, 3406–3415 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Basu, S. et al. “Emergency” granulopoiesis in G-CSF-deficient mice in response to Candida albicans infection. Blood 95, 3725–3733 (2000).

    Article  CAS  PubMed  Google Scholar 

  30. Manel, N. et al. A cryptic sensor for HIV-1 activates antiviral innate immunity in dendritic cells. Nature 467, 214–217 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Johanson, T.M., Lew, A.M. & Chong, M.M. MicroRNA-independent roles of the RNase III enzymes Drosha and Dicer. Open Biol. 3, 130144 (2013).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  32. Knuckles, P. et al. Drosha regulates neurogenesis by controlling neurogenin 2 expression independent of microRNAs. Nat. Neurosci. 15, 962–969 (2012).

    Article  CAS  PubMed  Google Scholar 

  33. Cheloufi, S., Dos Santos, C.O., Chong, M.M. & Hannon, G.J. A dicer-independent miRNA biogenesis pathway that requires Ago catalysis. Nature 465, 584–589 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Johanson, T.M. et al. A microRNA expression atlas of mouse dendritic cell development. Immunol. Cell Biol. 93, 480–485 (2015).

    Article  CAS  PubMed  Google Scholar 

  35. Shenoy, A. & Blelloch, R. Genomic analysis suggests that mRNA destabilization by the microprocessor is specialized for the auto-regulation of Dgcr8. PLoS ONE 4, e6971 (2009).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  36. Chong, M.M., Rasmussen, J.P., Rudensky, A.Y. & Littman, D.R. The RNAseIII enzyme Drosha is critical in T cells for preventing lethal inflammatory disease. J. Exp. Med. 205, 2005–2017 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Teta, M. et al. Inducible deletion of epidermal Dicer and Drosha reveals multiple functions for miRNAs in postnatal skin. Development 139, 1405–1416 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Zhdanova, O. et al. The inducible deletion of Drosha and microRNAs in mature podocytes results in a collapsing glomerulopathy. Kidney Int. 80, 719–730 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Gromak, N. et al. Drosha regulates gene expression independently of RNA cleavage function. Cell Rep. 5, 1499–1510 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Davis, B.N. & Hata, A. Regulation of microRNA biogenesis: a miRiad of mechanisms. Cell Commun. Signal. 7, 18 (2009).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  41. Newman, M.A., Thomson, J.M. & Hammond, S.M. Lin-28 interaction with the Let-7 precursor loop mediates regulated microRNA processing. RNA 14, 1539–1549 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Naik, S.H. et al. Intrasplenic steady-state dendritic cell precursors that are distinct from monocytes. Nat. Immunol. 7, 663–671 (2006).

    Article  CAS  PubMed  Google Scholar 

  43. Ding, Y. et al. FLT3-ligand treatment of humanized mice results in the generation of large numbers of CD141+ and CD1c+ dendritic cells in vivo. J. Immunol. 192, 1982–1989 (2014).

    Article  CAS  PubMed  Google Scholar 

  44. Lieschke, G.J. et al. Mice lacking granulocyte colony-stimulating factor have chronic neutropenia, granulocyte and macrophage progenitor cell deficiency, and impaired neutrophil mobilization. Blood 84, 1737–1746 (1994).

    Article  CAS  PubMed  Google Scholar 

  45. Smyth, G.K. Linear models and empirical Bayes methods for assessing differential expression in microarray experiments. Stat. Appl. Genet. Mol. Biol. 3, 3 (2004).

    Article  Google Scholar 

  46. Ritchie, M.E. et al. Empirical array quality weights in the analysis of microarray data. BMC Bioinformatics 7, 261 (2006).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  47. Yang, Y.H. et al. Normalization for cDNA microarray data: a robust composite method addressing single and multiple slide systematic variation. Nucleic Acids Res. 30, e15 (2002).

    Article  PubMed  PubMed Central  Google Scholar 

  48. Pear, W.S., Nolan, G.P., Scott, M.L. & Baltimore, D. Production of high-titer helper-free retroviruses by transient transfection. Proc. Natl. Acad. Sci. USA 90, 8392–8396 (1993).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank C. Yates for animal husbandry. Supported by the National Health and Medical Research Council, Australia (637338, 1004541, 1042211 and 1079586 to M.M.W.C.; 1037321, 1043414 and 1080321 to A.M.L.), the Juvenile Diabetes Research Foundation (5-2011-100 to M.M.W.C.), the Diabetes Australia Research Trust (Y13G-CHOM to M.M.W.C.), the Australian Research Council (M.M.W.C.) and the Victorian State Government Operational Infrastructure Support and Australian National Health and Medical Research Council Research Institute Infrastructure Support Scheme.

Author information

Authors and Affiliations

Authors

Contributions

T.M.J. performed most of the experiments with help from A.A.K., J.H.C.Y. and Y.Z.; M.C. and A.K. performed the bioinformatics analyses; and T.M.J., A.M.L., Y.Z. and M.M.W.C. designed the study, analyzed the data and wrote the manuscript.

Corresponding author

Correspondence to Mark M W Chong.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Integrated supplementary information

Supplementary Figure 1 Residual splenic DCs in Drosha-deficient chimeras have escaped deletion.

The residual splenic DCs in Tamoxifen-treated Droshafl/fl CreER chimeras were sorted by FACS, then Drosha mRNA levels were measured by quantitative RT-PCR.

Source data

Supplementary Figure 2 The accumulation of LSKs and CMPs caused by Drosha deficiency is cell intrinsic.

Comparison of LSK and CMP populations between the CD45.2 experimental and CD45.1 wild type supporting component in the bone marrow of chimeric mice following Tamoxifen treatment. The data shown is the mean +/- S.D. of twelve (control, Droshafl/fl CreER) or nine (Dicerfl/fl CreER) animals for each genotype from three independent experiments.

Source data

Supplementary Figure 3 Drosha ablation and miRNA expression are equivalent in LSK-, CMP-, CLP- and CDP-derived cultures.

Quantitative RT-PCR or Taqman-based quantitative RT-PCR determined levels of Drosha mRNA or a panel of mature miRNAs, respectively. The data shown is the mean +/- S.D. of one or two (LSK culture Drosha levels) independent experiments.

Source data

Supplementary Figure 4 Identification of cleavage sites within Myl9 mRNA and Todr1 mRNA.

(a) Schematic of 5’P capture experiment. PolyA transcripts were purified from total RNA isolated from lineage-depleted bone marrow cells. An adaptor was ligated specifically to transcripts exhibiting a 5’ monophosphate (a mark of endonucleolytic cleavage) and reverse transcribed. Tiled PCRs were then performed using a forward primer in the adaptor and reverse primers within Myl9 and Todr1 mRNA. Thus, PCR products could only be obtained if Myl9 and Todr1 contained a 5’ phosphate. PCR products were Sanger sequenced. The junction between the adaptor and mRNA denoted the original cleavage site. (b) Location of cleavage sites detected in Myl9 and Todr1 mRNA. Blue and red text denote stem or loop structure, respectively.

Supplementary Figure 5 Mutagenesis of the Todr1 stem-loop to disrupt base pairing.

Site-directed mutagenesis was used to introduce mismatch mutations into the sequence of the 5’ arm of the putative stem-loop structure within Todr1 mRNA.

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–5 and Supplementary Tables 1 and 2. (PDF 1482 kb)

Source data

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Johanson, T., Keown, A., Cmero, M. et al. Drosha controls dendritic cell development by cleaving messenger RNAs encoding inhibitors of myelopoiesis. Nat Immunol 16, 1134–1141 (2015). https://doi.org/10.1038/ni.3293

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/ni.3293

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