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.

  • Resource
  • Published:

An integrated expression atlas of miRNAs and their promoters in human and mouse

Abstract

MicroRNAs (miRNAs) are short non-coding RNAs with key roles in cellular regulation. As part of the fifth edition of the Functional Annotation of Mammalian Genome (FANTOM5) project, we created an integrated expression atlas of miRNAs and their promoters by deep-sequencing 492 short RNA (sRNA) libraries, with matching Cap Analysis Gene Expression (CAGE) data, from 396 human and 47 mouse RNA samples. Promoters were identified for 1,357 human and 804 mouse miRNAs and showed strong sequence conservation between species. We also found that primary and mature miRNA expression levels were correlated, allowing us to use the primary miRNA measurements as a proxy for mature miRNA levels in a total of 1,829 human and 1,029 mouse CAGE libraries. We thus provide a broad atlas of miRNA expression and promoters in primary mammalian cells, establishing a foundation for detailed analysis of miRNA expression patterns and transcriptional control regions.

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: Selection of robust miRNAs and Drosha CAGE peak analysis.
Figure 2: Expression profile and cell ontology analysis of mature miRNAs.
Figure 3: Analysis of the curated miRNA promoters of miRNAs in the robust set.

Similar content being viewed by others

Accession codes

Primary accessions

DDBJ/GenBank/EMBL

Gene Expression Omnibus

Referenced accessions

DDBJ/GenBank/EMBL

References

  1. Bartel, D.P. MicroRNAs: genomics, biogenesis, mechanism, and function. Cell 116, 281–297 (2004).

    Article  CAS  PubMed  Google Scholar 

  2. Shenoy, A. & Blelloch, R.H. Regulation of microRNA function in somatic stem cell proliferation and differentiation. Nat. Rev. Mol. Cell Biol. 15, 565–576 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  3. Li, M. & Izpisua Belmonte, J.C. Roles for noncoding RNAs in cell-fate determination and regeneration. Nat. Struct. Mol. Biol. 22, 2–4 (2015).

    CAS  PubMed  Google Scholar 

  4. Mehta, A. & Baltimore, D. MicroRNAs as regulatory elements in immune system logic. Nat. Rev. Immunol. 16, 279–294 (2016).

    CAS  PubMed  Google Scholar 

  5. Hasuwa, H., Ueda, J., Ikawa, M. & Okabe, M. miR-200b and miR-429 function in mouse ovulation and are essential for female fertility. Science 341, 71–73 (2013).

    CAS  PubMed  Google Scholar 

  6. Sun, K. & Lai, E.C. Adult-specific functions of animal microRNAs. Nat. Rev. Genet. 14, 535–548 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  7. Mendell, J.T. & Olson, E.N. MicroRNAs in stress signaling and human disease. Cell 148, 1172–1187 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  8. Adams, B.D., Kasinski, A.L. & Slack, F.J. Aberrant regulation and function of microRNAs in cancer. Curr. Biol. 24, R762–R776 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  9. Lin, S. & Gregory, R.I. MicroRNA biogenesis pathways in cancer. Nat. Rev. Cancer 15, 321–333 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  10. Jonas, S. & Izaurralde, E. Towards a molecular understanding of microRNA-mediated gene silencing. Nat. Rev. Genet. 16, 421–433 (2015).

    CAS  PubMed  Google Scholar 

  11. Ha, M. & Kim, V.N. Regulation of microRNA biogenesis. Nat. Rev. Mol. Cell Biol. 15, 509–524 (2014).

    CAS  PubMed  Google Scholar 

  12. Kozomara, A. & Griffiths-Jones, S. miRBase: annotating high confidence microRNAs using deep sequencing data. Nucleic Acids Res. 42, D68–D73 (2014).

    CAS  PubMed  Google Scholar 

  13. Pritchard, C.C., Cheng, H.H. & Tewari, M. MicroRNA profiling: approaches and considerations. Nat. Rev. Genet. 13, 358–369 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  14. Chang, T.C., Pertea, M., Lee, S., Salzberg, S.L. & Mendell, J.T. Genome-wide annotation of microRNA primary transcript structures reveals novel regulatory mechanisms. Genome Res. 25, 1401–1409 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  15. Kanamori-Katayama, M. et al. Unamplified cap analysis of gene expression on a single-molecule sequencer. Genome Res. 21, 1150–1159 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  16. Takahashi, H., Lassmann, T., Murata, M. & Carninci, P. 5′ end-centered expression profiling using cap-analysis gene expression and next-generation sequencing. Nat. Protoc. 7, 542–561 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  17. Forrest, A.R.R. et al. A promoter-level mammalian expression atlas. Nature 507, 462–470 (2014).

    CAS  PubMed  Google Scholar 

  18. Arner, E. et al. Transcribed enhancers lead waves of coordinated transcription in transitioning mammalian cells. Science 347, 1010–1014 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  19. Fort, A. et al. Deep transcriptome profiling of mammalian stem cells supports a regulatory role for retrotransposons in pluripotency maintenance. Nat. Genet. 46, 558–566 (2014).

    CAS  PubMed  Google Scholar 

  20. Fromm, B. et al. A uniform system for the annotation of vertebrate microRNA genes and the evolution of the human microRNAome. Annu. Rev. Genet. 49, 213–242 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  21. Nepal, C. et al. Transcriptional, post-transcriptional and chromatin-associated regulation of pri-miRNAs, pre-miRNAs and moRNAs. Nucleic Acids Res. 44, 3070–3081 (2016).

    CAS  PubMed  Google Scholar 

  22. Djebali, S. et al. Landscape of transcription in human cells. Nature 489, 101–108 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  23. Taft, R.J. et al. Tiny RNAs associated with transcription start sites in animals. Nat. Genet. 41, 572–578 (2009).

    CAS  PubMed  Google Scholar 

  24. Westholm, J.O. & Lai, E.C. Mirtrons: microRNA biogenesis via splicing. Biochimie 93, 1897–1904 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  25. Matera, A.G., Terns, R.M. & Terns, M.P. Non-coding RNAs: lessons from the small nuclear and small nucleolar RNAs. Nat. Rev. Mol. Cell Biol. 8, 209–220 (2007).

    CAS  PubMed  Google Scholar 

  26. Friedländer, M.R., Mackowiak, S.D., Li, N., Chen, W. & Rajewsky, N. miRDeep2 accurately identifies known and hundreds of novel microRNA genes in seven animal clades. Nucleic Acids Res. 40, 37–52 (2012).

    PubMed  Google Scholar 

  27. Londin, E. et al. Analysis of 13 cell types reveals evidence for the expression of numerous novel primate- and tissue-specific microRNAs. Proc. Natl. Acad. Sci. USA 112, E1106–E1115 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  28. Freeman, T.C. et al. Construction, visualisation, and clustering of transcription networks from microarray expression data. PLOS Comput. Biol. 3, 2032–2042 (2007).

    CAS  PubMed  Google Scholar 

  29. Ludwig, N. et al. Distribution of miRNA expression across human tissues. Nucleic Acids Res. 44, 3865–3877 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  30. Meehan, T.F. et al. Logical development of the cell ontology. BMC Bioinformatics 12, 6 (2011).

    PubMed  PubMed Central  Google Scholar 

  31. Lizio, M. et al. Gateways to the FANTOM5 promoter level mammalian expression atlas. Genome Biol. 16, 22 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  32. Batut, P., Dobin, A., Plessy, C., Carninci, P. & Gingeras, T.R. High-fidelity promoter profiling reveals widespread alternative promoter usage and transposon-driven developmental gene expression. Genome Res. 23, 169–180 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  33. Georgakilas, G. et al. DIANA-miRGen v3.0: accurate characterization of microRNA promoters and their regulators. Nucleic Acids Res. 44 D1, D190–D195 (2016).

    CAS  PubMed  Google Scholar 

  34. Chien, C.H. et al. Identifying transcriptional start sites of human microRNAs based on high-throughput sequencing data. Nucleic Acids Res. 39, 9345–9356 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  35. Guo, Z. et al. Genome-wide survey of tissue-specific microRNA and transcription factor regulatory networks in 12 tissues. Sci. Rep. 4, 5150 (2014).

    PubMed  PubMed Central  Google Scholar 

  36. Ding, J. et al. Trbp regulates heart function through microRNA-mediated Sox6 repression. Nat. Genet. 47, 776–783 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  37. Suzuki, H. et al. The transcriptional network that controls growth arrest and differentiation in a human myeloid leukemia cell line. Nat. Genet. 41, 553–562 (2009).

    CAS  PubMed  Google Scholar 

  38. Landgraf, P. et al. A mammalian microRNA expression atlas based on small RNA library sequencing. Cell 129, 1401–1414 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  39. Carninci, P. et al. The transcriptional landscape of the mammalian genome. Science 309, 1559–1563 (2005).

    CAS  PubMed  Google Scholar 

  40. Kapranov, P. et al. RNA maps reveal new RNA classes and a possible function for pervasive transcription. Science 316, 1484–1488 (2007).

    CAS  PubMed  Google Scholar 

  41. Meunier, J. et al. Birth and expression evolution of mammalian microRNA genes. Genome Res. 23, 34–45 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  42. Francia, S. et al. Site-specific DICER and DROSHA RNA products control the DNA-damage response. Nature 488, 231–235 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  43. Valen, E. et al. Biogenic mechanisms and utilization of small RNAs derived from human protein-coding genes. Nat. Struct. Mol. Biol. 18, 1075–1082 (2011).

    CAS  PubMed  Google Scholar 

  44. Siepel, A. et al. Evolutionarily conserved elements in vertebrate, insect, worm, and yeast genomes. Genome Res. 15, 1034–1050 (2005).

    CAS  PubMed  PubMed Central  Google Scholar 

  45. Abugessaisa, I. et al. FANTOM5 transcriptome catalog of cellular states based on Semantic MediaWiki. Database (Oxford) 2016, baw105 (2016).

    Google Scholar 

  46. Lassmann, T., Hayashizaki, Y. & Daub, C.O. TagDust—a program to eliminate artifacts from next generation sequencing data. Bioinformatics 25, 2839–2840 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  47. Li, H. & Durbin, R. Fast and accurate short read alignment with Burrows-Wheeler transform. Bioinformatics 25, 1754–1760 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  48. De Hoon, M.J.L. et al. Cross-mapping and the identification of editing sites in mature microRNAs in high-throughput sequencing libraries. Genome Res. 20, 257–264 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  49. Lorenz, R. et al. ViennaRNA Package 2.0. Algorithms Mol. Biol. 6, 26 (2011).

    PubMed  PubMed Central  Google Scholar 

  50. Vijayan, D., Radford, K.J., Beckhouse, A.G., Ashman, R.B. & Wells, C.A. Mincle polarizes human monocyte and neutrophil responses to Candida albicans. Immunol. Cell Biol. 90, 889–895 (2012).

    CAS  PubMed  Google Scholar 

  51. Busk, P.K. A tool for design of primers for microRNA-specific quantitative RT-qPCR. BMC Bioinformatics 15, 29 (2014).

    PubMed  PubMed Central  Google Scholar 

  52. Schwarzenbach, H., da Silva, A.M., Calin, G. & Pantel, K. Data normalization strategies for microRNA quantification. Clin. Chem. 61, 1333–1342 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  53. Nagpal, N. & Kulshreshtha, R. miR-191: an emerging player in disease biology. Front. Genet. 5, 99 (2014).

    PubMed  PubMed Central  Google Scholar 

  54. Moon, H.G., Yang, J., Zheng, Y. & Jin, Y. miR-15a/16 regulates macrophage phagocytosis after bacterial infection. J. Immunol. 193, 4558–4567 (2014).

    CAS  PubMed  Google Scholar 

  55. Vinod, M. et al. miR-206 controls LXRα expression and promotes LXR-mediated cholesterol efflux in macrophages. Biochim. Biophys. Acta 1841, 827–835 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  56. Cobos Jiménez, V. et al. Next-generation sequencing of microRNAs in primary human polarized macrophages. Genom. Data 2, 181–183 (2014).

    PubMed  PubMed Central  Google Scholar 

  57. Zhang, L. et al. miR-153 supports colorectal cancer progression via pleiotropic effects that enhance invasion and chemotherapeutic resistance. Cancer Res. 73, 6435–6447 (2013).

    CAS  PubMed  Google Scholar 

  58. Srivastava, S.K. et al. MicroRNA-345 induces apoptosis in pancreatic cancer cells through potentiation of caspase-dependent and -independent pathways. Br. J. Cancer 113, 660–668 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  59. Robinson, M.D., McCarthy, D.J. & Smyth, G.K. edgeR: a Bioconductor package for differential expression analysis of digital gene expression data. Bioinformatics 26, 139–140 (2010).

    CAS  PubMed  Google Scholar 

  60. Anders, S. & Huber, W. Differential expression analysis for sequence count data. Genome Biol. 11, R106 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  61. Harrow, J. et al. GENCODE: the reference human genome annotation for The ENCODE Project. Genome Res. 22, 1760–1774 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  62. Brown, G.R. et al. Gene: a gene-centered information resource at NCBI. Nucleic Acids Res. 43, D36–D42 (2015).

    CAS  PubMed  Google Scholar 

  63. Kent, W.J. BLAT—the BLAST-like alignment tool. Genome Res. 12, 656–664 (2002).

    CAS  PubMed  PubMed Central  Google Scholar 

  64. Karolchik, D. et al. The UCSC Genome Browser database: 2014 update. Nucleic Acids Res. 42, D764–D770 (2014).

    CAS  PubMed  Google Scholar 

  65. Notredame, C., Higgins, D.G. & Heringa, J. T-Coffee: A novel method for fast and accurate multiple sequence alignment. J. Mol. Biol. 302, 205–217 (2000).

    CAS  PubMed  Google Scholar 

  66. Arnold, P., Erb, I., Pachkov, M., Molina, N. & van Nimwegen, E. MotEvo: integrated Bayesian probabilistic methods for inferring regulatory sites and motifs on multiple alignments of DNA sequences. Bioinformatics 28, 487–494 (2012).

    CAS  PubMed  Google Scholar 

  67. Pachkov, M., Balwierz, P.J., Arnold, P., Ozonov, E. & van Nimwegen, E. SwissRegulon, a database of genome-wide annotations of regulatory sites: recent updates. Nucleic Acids Res. 41, D214–D220 (2013).

    CAS  PubMed  Google Scholar 

Download references

Acknowledgements

FANTOM5 was made possible by the following grants: Research Grant for RIKEN Omics Science Center from MEXT to Y.H.; Grant of the Innovative Cell Biology by Innovative Technology (Cell Innovation Program) from the MEXT to Y.H.; Research Grant from MEXT to the RIKEN Center for Life Science Technologies; Research Grant to RIKEN Preventive Medicine and Diagnosis Innovation Program from MEXT to Y.H. K.V.-S. and A.S. were supported by the Lundbeck and Novo Nordisk Foundations. A.R.R.F. is supported by a Senior Cancer Research Fellowship from the Cancer Research Trust, funds raised by the MACA Ride to Conquer Cancer, and the Australian Research Council's Discovery Projects funding scheme (DP160101960). Y.A.M. was supported by the Russian Science Foundation, grant 15-14-30002. R.D. was supported by the Russian Science Foundation, grant 14-44-00022. We would like to thank L. Schwarzfischer for technical assistance and N. Eichner and G. Meister for sequencing RACE products. We would also like to thank GeNAS for data production.

Author information

Authors and Affiliations

Authors

Consortia

Contributions

P.A., G.Å., M.B., A.J.C., M.D., D.G., S.G., T.J.H., M.H., P.H., K.J.H., C.K., P.K., W.L., N.M., M.O., M.O.-H., P.R., H.S., R.K.S., H.To., M.Y., N.Y., S.Z., P.G.Z., L.W., Y.Y., C.A.W., K.M.S., and A.R.R.F. provided RNA samples; E.A. and C.O.D. selected samples from the FANTOM5 time courses; Y.I., S.N., and H.Ta. produced the sRNA libraries; I.A., M.L., H.K., and T.K. managed the data; D.d.R., M.J.L.d.H., K.V.-S., A.M.B., T.A., H.A., A.H., T.L., H.P., C.-H.L. A.M., V.M., and M.R. carried out the bioinformatics analyses with the help of C.C.H., M.L., K.H., F.R., and J.S.; C.J.M. provided the cell ontology; K.M.S. created the Miru visualization; A.F., A.M., A.R.R.F., A.S., C.-H.L. C.A.W., D.d.R., E.H., F.R., H.P., K.V.-S., A.M.B., M.J.L.d.H., M.R., N.B., P.S., R.D., V.M., and Y.A.M. contributed to the manual miRNA promoter annotation; K.Y. and J.W.S. performed the expression validation experiments of known miRNAs; E.H. and C.A.W. performed the validation experiments of candidate miRNAs; C.G. and M.R. performed the RACE experiments; J.H. created the web visualization tool; D.d.R., A.R.R.F., and M.J.L.d.H. wrote the manuscript with the help of E.A., A.S., A.M.B., K.M.S., K.V.-S., M.R., N.B., P.C., P.S., and C.A.W.; A.R.R.F. and M.J.L.d.H. designed the study; P.C. and Y.H. supervised the FANTOM5 project.

Corresponding authors

Correspondence to Alistair R R Forrest or Michiel J L de Hoon.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–29 and Supplementary Note (PDF 3223 kb)

Life Sciences Reporting Summary (PDF 158 kb)

Supplementary Table 1

Short RNA data sets analyzed in this study. (XLSX 37 kb)

Supplementary Table 2

Novel RNA samples used. Most FANTOM5 human and mouse RNA samples used were described previously (ref. 17,18) and are therefore not included in this table. (XLSX 55 kb)

Supplementary Table 3

FANTOM5 RNA samples and sRNA libraries. Matching CAGE (ref. 17–19) and sRNA libraries were produced from the same RNA sample. In total, five of the CAGE libraries and two of the sRNA libraries were discarded because of their low quality; for one of the RNA samples, an sRNA library but no CAGE library was produced. (XLSX 38 kb)

Supplementary Table 4

Evaluation of human pre-miRNAs. For each pre-miRNA in the human robust, permissive, and candidate set, we evaluated the miRBase high-confidence criteria (Table 2), and the statistical significance of the Drosha CAGE peak as observed in the FANTOM5 and ENCODE CAGE data. (XLSX 463 kb)

Supplementary Table 5

Evaluation of murine pre-miRNAs. For each pre-miRNA in the murine robust, permissive, and candidate set, we evaluated the miRBase high-confidence criteria (Table 2), and the statistical significance of the Drosha CAGE peak as observed in the FANTOM5 CAGE data. (XLSX 161 kb)

Supplementary Table 6

Genomic locations of the candidate miRNAs predicted by miRDeep2 in human (genome assembly hg19). (XLSX 764 kb)

Supplementary Table 7

Genome sequence at the genomic locus of each candidate miRNA in human, the secondary structure of the predicted pre-miRNA with the corresponding ΔG, and aligning reads with their counts. Sequenced nucleotides that do not match the genome sequence are shown in lowercase. (XLSX 2535 kb)

Supplementary Table 8

Genomic locations of the candidate miRNAs predicted by miRDeep2 in mouse (genome assembly mm9). (XLSX 215 kb)

Supplementary Table 9

Genome sequence at the genomic locus of each candidate miRNA in mouse, the secondary structure of the predicted pre-miRNA with the corresponding ΔG, and aligning reads with their counts. Sequenced nucleotides that do not match the genome sequence are shown in lowercase. (XLSX 704 kb)

Supplementary Table 10

Forward primers used for the validation of candidate miRNA expression by qPCR. (XLSX 41 kb)

Supplementary Table 11

Expression table of human miRNAs in the robust, permissive, and candidate set. The values shown are the (unnormalized) counts of sequence reads overlapping the mature miRNA region, and may be non-integer due to sequence reads mapping to multiple genomic locations. (XLSX 19326 kb)

Supplementary Table 12

Expression table of murine miRNAs in the robust, permissive, and candidate set. The values shown are the (unnormalized) counts of sequence reads overlapping the mature miRNA region, and may be non-integer due to sequence reads mapping to multiple genomic locations. (XLSX 1334 kb)

Supplementary Table 13

Cell ontology enrichment analysis. For each mature miRNA, we show the cell type specificity index, the median and maximum expression level, the RNA sample in which the miRNA was most highly expressed, the top-3 cell ontology clusters in which its expression is most enriched, with the corresponding significance value and the base-2 logarithm of the expression fold-ratio, and the top-3 cell ontology clusters in which its expression is most depleted, with the corresponding significance value and the base-2 logarithm of the expression fold-ratio. (XLSX 974 kb)

Supplementary Table 14

RNA samples contained in each cell ontology cluster (sRNA data). (XLSX 37 kb)

Supplementary Table 15

Computational miRNA promoter predictions in human. For each primary miRNA, we show the genomic location (chromosome, strand, and transcription start site; genome assembly hg19) and name of the predicted promoter, the corresponding primary miRNA, their status as intronic (if the primary miRNA transcript is coding) or intergenic (if the primary miRNA is non-coding), the pre-miRNAs contained in the primary miRNA, the average sequence conservation of the miRNA promoter, the maximum CAGE expression level, the RNA sample in which the primary miRNA promoter was most highly expressed, the top-3 cell ontology clusters in which CAGE expression of this promoter is most enriched, with the corresponding statistical significance and the base-2 logarithm of the expression fold-ratio, and the top-3 cell ontology clusters in which CAGE expression of this promoter is most depleted, with the corresponding statistical significance and the base-2 logarithm of the expression fold-ratio. The promoter loci and names were taken from the FANTOM5 permissive promoter set (ref. 17). (XLSX 1121 kb)

Supplementary Table 16

MicroRNA promoter predictions in mouse. For each primary miRNA, we show the genomic location (chromosome, strand, and transcription start site; genome assembly mm9) and name of the predicted promoter, the corresponding primary miRNA, their status as intronic (if the primary miRNA transcript is coding) or intergenic (if the primary miRNA is non-coding), the pre-miRNAs contained in the primary miRNA, and the average sequence conservation of the miRNA promoter. The promoter loci and names were taken from the FANTOM5 permissive promoter set (ref. 17). (XLSX 165 kb)

Supplementary Table 17

Curated miRNA promoter predictions in human. For each primary miRNA, we show the genomic location (chromosome, strand, and transcription start site; genome assembly hg19) and name of the predicted promoter, the corresponding primary miRNA, their status as intronic (if the primary miRNA transcript is coding) or intergenic (if the primary miRNA is non-coding), the pre-miRNAs contained in the primary miRNA, the average sequence conservation of the miRNA promoter, the maximum CAGE expression level, the RNA sample in which the primary miRNA promoter was most highly expressed, the top-3 cell ontology clusters in which CAGE expression of this promoter is most enriched, with the corresponding statistical significance and the base-2 logarithm of the expression fold-ratio, and the top-3 cell ontology clusters in which CAGE expression of this promoter is most depleted, with the corresponding statistical significance and the base-2 logarithm of the expression fold-ratio. The promoter loci and names were taken from the FANTOM5 permissive promoter set (ref. 17). (XLSX 1145 kb)

Supplementary Table 18

Outer and inner primers used for the validation of miRNA promoters by RACE. (XLSX 35 kb)

Supplementary Table 19

Spearman correlation across human primary cells between the mature miRNA expression, as measured by sRNA sequencing, and the miRNA promoter, as measured by CAGE. (XLSX 242 kb)

Supplementary Table 20

RNA samples contained in each cell ontology cluster (CAGE data). (XLSX 76 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

de Rie, D., Abugessaisa, I., Alam, T. et al. An integrated expression atlas of miRNAs and their promoters in human and mouse. Nat Biotechnol 35, 872–878 (2017). https://doi.org/10.1038/nbt.3947

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nbt.3947

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