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:

The RNA N6-methyladenosine modification landscape of human fetal tissues

Abstract

A single genome gives rise to diverse tissues through complex epigenomic mechanisms, including N6-methyladenosine (m6A), a widespread RNA modification that is implicated in many biological processes. Here, to explore the global landscape of m6A in human tissues, we generated 21 whole-transcriptome m6A methylomes across major fetal tissues using m6A sequencing. These data reveal dynamic m6A methylation, identify large numbers of tissue differential m6A modifications and indicate that m6A is positively correlated with gene expression homeostasis. We also report m6A methylomes of long intergenic non-coding RNA (lincRNA), finding that enhancer lincRNAs are enriched for m6A. Tissue m6A regions are often enriched for single nucleotide polymorphisms that are associated with the expression of quantitative traits and complex traits including common diseases, which may potentially affect m6A modifications. Finally, we find that m6A modifications preferentially occupy genes with CpG-rich promoters, features of which regulate RNA transcript m6A. Our data indicate that m6A is widely regulated by human genetic variation and promoters, suggesting a broad involvement of m6A in human development and disease.

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

Fig. 1: Whole-transcriptome profiling of m6As in major human tissue types.
Fig. 2: Differential m6A methylation among human tissue types.
Fig. 3: The m6A methylation landscape of human lincRNAs.
Fig. 4: m6A methylation is correlated with gene expression homeostasis across human tissues.
Fig. 5: Tissue m6As exhibit increased eQTL SNP frequency and enrichment in non-random GWAS SNPs.
Fig. 6: m6As are enriched in genes with CpG-rich promoters.
Fig. 7: The CpG status of promoters is involved in m6A methylation.

Similar content being viewed by others

Data availability

MeRIP–seq, input RNA-seq and ChIP–seq data that support the findings of this study have been deposited in the Gene Expression Omnibus under accession code GSE114150. Source data for Figs. 17 and Supplementary Figs. 17 have been provided as Supplementary Table 5. All other data supporting the findings of this study are available from the corresponding author on reasonable request.

Code availability

Custom codes were written in Python and R based on published software or papers, and are available on GitHub (https://github.com/XiaLabBioinformatics/m6AMethylation/).

References

  1. Dominissini, D. et al. Topology of the human and mouse m6A RNA methylomes revealed by m6A-seq. Nature 485, 201–206 (2012).

    Article  CAS  Google Scholar 

  2. Meyer, K. D. et al. Comprehensive analysis of mRNA methylation reveals enrichment in 3’ UTRs and near stop codons. Cell 149, 1635–1646 (2012).

    Article  CAS  Google Scholar 

  3. Zhao, B. S., Roundtree, I. A. & He, C. Post-transcriptional gene regulation by mRNA modifications. Nat. Rev. Mol. Cell Biol. 18, 31–42 (2017).

    Article  CAS  Google Scholar 

  4. Liu, J. et al. A METTL3–METTL14 complex mediates mammalian nuclear RNA N 6-adenosine methylation. Nat. Chem. Biol. 10, 93–95 (2014).

    Article  CAS  Google Scholar 

  5. Jia, G. et al. N6-Methyladenosine in nuclear RNA is a major substrate of the obesity-associated FTO. Nat. Chem. Biol. 7, 885–887 (2011).

    Article  CAS  Google Scholar 

  6. Zheng, G. et al. ALKBH5 is a mammalian RNA demethylase that impacts RNA metabolism and mouse fertility. Mol. Cell 49, 18–29 (2013).

    Article  CAS  Google Scholar 

  7. Roundtree, I. A., Evans, M. E., Pan, T. & He, C. Dynamic RNA modifications in gene expression regulation. Cell 169, 1187–1200 (2017).

    Article  CAS  Google Scholar 

  8. Wang, X. et al. N6-Methyladenosine-dependent regulation of messenger RNA stability. Nature 505, 117–120 (2014).

    Article  Google Scholar 

  9. Huang, H. et al. Recognition of RNA N 6-methyladenosine by IGF2BP proteins enhances mRNA stability and translation. Nat. Cell Biol. 20, 285–295 (2018).

    Article  CAS  Google Scholar 

  10. Alarcon, C. R. et al. HNRNPA2B1 is a mediator of m6A-dependent nuclear RNA processing events. Cell 162, 1299–1308 (2015).

    Article  CAS  Google Scholar 

  11. Liu, N. et al. N6-methyladenosine-dependent RNA structural switches regulate RNA–protein interactions. Nature 518, 560–564 (2015).

    Article  CAS  Google Scholar 

  12. Ke, S. et al. m6A mRNA modifications are deposited in nascent pre-mRNA and are not required for splicing but do specify cytoplasmic turnover. Genes Dev. 31, 990–1006 (2017).

    Article  CAS  Google Scholar 

  13. Xiao, W. et al. Nuclear m6A reader YTHDC1 regulates mRNA splicing. Mol. Cell 61, 507–519 (2016).

    Article  CAS  Google Scholar 

  14. Pendleton, K. E. et al. The U6 snRNA m6A methyltransferase METTL16 regulates SAM synthetase intron retention. Cell 169, 824–835 (2017).

    Article  CAS  Google Scholar 

  15. Wang, X. et al. N6-Methyladenosine modulates messenger RNA translation efficiency. Cell 161, 1388–1399 (2015).

    Article  CAS  Google Scholar 

  16. Zhou, J. et al. Dynamic m6A mRNA methylation directs translational control of heat shock response. Nature 526, 591–594 (2015).

    Article  CAS  Google Scholar 

  17. Meyer, K. D. et al. 5’ UTR m6A promotes cap-independent translation. Cell 163, 999–1010 (2015).

    Article  CAS  Google Scholar 

  18. Shi, H. et al. YTHDF3 facilitates translation and decay of N 6-methyladenosine-modified RNA. Cell Res. 27, 315–328 (2017).

    Article  CAS  Google Scholar 

  19. Alarcon, C. R., Lee, H., Goodarzi, H., Halberg, N. & Tavazoie, S. F. N6-Methyladenosine marks primary microRNAs for processing. Nature 519, 482–485 (2015).

    Article  CAS  Google Scholar 

  20. Xiang, Y. et al. RNA m6A methylation regulates the ultraviolet-induced DNA damage response. Nature 543, 573–576 (2017).

    Article  CAS  Google Scholar 

  21. Wang, Y. et al. N6-Methyladenosine modification destabilizes developmental regulators in embryonic stem cells. Nat. Cell Biol. 16, 191–198 (2014).

    Article  CAS  Google Scholar 

  22. Batista, P. J. et al. m6A RNA modification controls cell fate transition in mammalian embryonic stem cells. Cell Stem Cell 15, 707–719 (2014).

    Article  CAS  Google Scholar 

  23. Geula, S. et al. m6A mRNA methylation facilitates resolution of naive pluripotency toward differentiation. Science 347, 1002–1006 (2015).

    Article  CAS  Google Scholar 

  24. Chen, T. et al. m6A RNA methylation is regulated by microRNAs and promotes reprogramming to pluripotency. Cell Stem Cell 16, 289–301 (2015).

    Article  CAS  Google Scholar 

  25. Zhang, C. et al. m6A modulates haematopoietic stem and progenitor cell specification. Nature 549, 273–276 (2017).

    Article  CAS  Google Scholar 

  26. Weng, H. et al. METTL14 inhibits hematopoietic stem/progenitor differentiation and promotes leukemogenesis via mRNA m6A modification. Cell Stem Cell 22, 191–205 (2018).

    Article  CAS  Google Scholar 

  27. Xu, K. et al. Mettl3-mediated m6A regulates spermatogonial differentiation and meiosis initiation. Cell Res. 27, 1100–1114 (2017).

    Article  CAS  Google Scholar 

  28. Haussmann, I. U. et al. m6A potentiates Sxl alternative pre-mRNA splicing for robust Drosophila sex determination. Nature 540, 301–304 (2016).

    Article  CAS  Google Scholar 

  29. Lence, T. et al. m6A modulates neuronal functions and sex determination in Drosophila. Nature 540, 242–247 (2016).

    Article  CAS  Google Scholar 

  30. Zhao, B. S. et al. m6A-dependent maternal mRNA clearance facilitates zebrafish maternal-to-zygotic transition. Nature 542, 475–478 (2017).

    Article  CAS  Google Scholar 

  31. Yoon, K. J. et al. Temporal control of mammalian cortical neurogenesis by m6A methylation. Cell 171, 877–889 (2017).

    Article  CAS  Google Scholar 

  32. Li, H. B. et al. m6A mRNA methylation controls T cell homeostasis by targeting the IL-7/STAT5/SOCS pathways. Nature 548, 338–342 (2017).

    Article  CAS  Google Scholar 

  33. Vu, L. P. et al. The N 6-methyladenosine (m6A)-forming enzyme METTL3 controls myeloid differentiation of normal hematopoietic and leukemia cells. Nat. Med. 23, 1369–1376 (2017).

    Article  CAS  Google Scholar 

  34. Zhang, C. et al. Hypoxia induces the breast cancer stem cell phenotype by HIF-dependent and ALKBH5-mediated m6A-demethylation of NANOG mRNA. Proc. Natl Acad. Sci. USA 113, E2047–E2056 (2016).

    Article  CAS  Google Scholar 

  35. Li, Z. et al. FTO plays an oncogenic role in acute myeloid leukemia as a N 6-methyladenosine RNA demethylase. Cancer Cell 31, 127–141 (2017).

    Article  Google Scholar 

  36. Su, R. et al. R-2HG exhibits anti-tumor activity by targeting FTO/m6A/MYC/CEBPA signaling. Cell 172, 90–105 (2018).

    Article  CAS  Google Scholar 

  37. Sun, W. J. et al. RMBase: a resource for decoding the landscape of RNA modifications from high-throughput sequencing data. Nucleic Acids Res. 44, D259–D265 (2016).

    Article  CAS  Google Scholar 

  38. Ransohoff, J. D., Wei, Y. & Khavari, P. A. The functions and unique features of long intergenic non-coding RNA. Nat. Rev. Mol. Cell Biol. 19, 143–157 (2018).

    Article  CAS  Google Scholar 

  39. Tilgner, H. et al. Deep sequencing of subcellular RNA fractions shows splicing to be predominantly co-transcriptional in the human genome but inefficient for lncRNAs. Genome Res. 22, 1616–1625 (2012).

    Article  CAS  Google Scholar 

  40. Hon, C. C. et al. An atlas of human long non-coding RNAs with accurate 5′ ends. Nature 543, 199–204 (2017).

    Article  CAS  Google Scholar 

  41. Majewski, J. & Pastinen, T. The study of eQTL variations by RNA-seq: from SNPs to phenotypes. Trend. Genet. 27, 72–79 (2011).

    Article  CAS  Google Scholar 

  42. Li, K. L. et al. Interaction between erythrocyte phospholipid fatty acids composition and variants of inflammation-related genes on type 2 diabetes. J. Nutrigenet. Nutrigenom. 7, 252–263 (2014).

    Article  CAS  Google Scholar 

  43. Frayling, T. M. et al. A common variant in the FTO gene is associated with body mass index and predisposes to childhood and adult obesity. Science 316, 889–894 (2007).

    Article  CAS  Google Scholar 

  44. Scott, L. J. et al. A genome-wide association study of type 2 diabetes in Finns detects multiple susceptibility variants. Science 316, 1341–1345 (2007).

    Article  CAS  Google Scholar 

  45. Slobodin, B. et al. Transcription impacts the efficiency of mRNA translation via co-transcriptional N6-adenosine methylation. Cell 169, 326–337 (2017).

    Article  CAS  Google Scholar 

  46. Knuckles, P. et al. RNA fate determination through cotranscriptional adenosine methylation and microprocessor binding. Nat. Struct. Mol. Biol. 24, 561–569 (2017).

    Article  CAS  Google Scholar 

  47. Barbieri, I. et al. Promoter-bound METTL3 maintains myeloid leukaemia by m6A-dependent translation control. Nature 552, 126–131 (2017).

    Article  CAS  Google Scholar 

  48. Bertero, A. et al. The SMAD2/3 interactome reveals that TGFβ controls m6A mRNA methylation in pluripotency. Nature 555, 256–259 (2018).

    Article  CAS  Google Scholar 

  49. The GTEx Consortium The Genotype-Tissue Expression (GTEx) pilot analysis: multitissue gene regulation in humans. Science 348, 648–660 (2015).

  50. Bergman, Y. & Cedar, H. DNA methylation dynamics in health and disease. Nat. Struct. Mol. Biol. 20, 274–281 (2013).

    Article  CAS  Google Scholar 

  51. Zid, B. M. & O’Shea, E. K. Promoter sequences direct cytoplasmic localization and translation of mRNAs during starvation in yeast. Nature 514, 117–121 (2014).

    Article  CAS  Google Scholar 

  52. Bregman, A. et al. Promoter elements regulate cytoplasmic mRNA decay. Cell 147, 1473–1483 (2011).

    Article  CAS  Google Scholar 

  53. Cramer, P. et al. Coupling of transcription with alternative splicing: RNA pol II promoters modulate SF2/ASF and 9G8 effects on an exonic splicing enhancer. Mol. Cell 4, 251–258 (1999).

    Article  CAS  Google Scholar 

  54. Wan, L. et al. Scaffolding protein SPIDR/KIAA0146 connects the Bloom syndrome helicase with homologous recombination repair. Proc. Natl Acad. Sci. USA 110, 10646–10651 (2013).

    Article  CAS  Google Scholar 

  55. Dominissini, D., Moshitch-Moshkovitz, S., Salmon-Divon, M., Amariglio, N. & Rechavi, G. Transcriptome-wide mapping of N 6-methyladenosine by m6A-seq based on immunocapturing and massively parallel sequencing. Nat. Protoc. 8, 176–189 (2013).

    Article  CAS  Google Scholar 

  56. Wan, Y. et al. Transcriptome-wide high-throughput deep m6A-seq reveals unique differential m6A methylation patterns between three organs in Arabidopsis thaliana. Genome Biol. 16, 272 (2015).

    Article  Google Scholar 

  57. Pertea, M., Kim, D., Pertea, G. M., Leek, J. T. & Salzberg, S. L. Transcript-level expression analysis of RNA-seq experiments with HISAT, StringTie and Ballgown. Nat. Protoc. 11, 1650–1667 (2016).

    Article  CAS  Google Scholar 

  58. Heinz, S. et al. Simple combinations of lineage-determining transcription factors prime cis-regulatory elements required for macrophage and B cell identities. Mol. Cell 38, 576–589 (2010).

    Article  CAS  Google Scholar 

  59. Cui, X. et al. Guitar: an R/Bioconductor package for gene annotation guided transcriptomic analysis of RNA-related genomic features. Biomed. Res. Int. 2016, 8367534 (2016).

    PubMed  PubMed Central  Google Scholar 

  60. Wang, L. et al. Measure transcript integrity using RNA-seq data. BMC Bioinform. 17, 58 (2016).

    Article  Google Scholar 

  61. Ke, S. et al. m6A mRNA modifications are deposited in nascent pre-mRNA and are not required for splicing but do specify cytoplasmic turnover. Genes Dev. 31, 990–1006 (2017).

    Article  CAS  Google Scholar 

  62. Shen, S. et al. rMATS: robust and flexible detection of differential alternative splicing from replicate RNA-seq data. Proc. Natl Acad. Sci. USA 111, E5593–E5601 (2014).

    Article  CAS  Google Scholar 

  63. Roadmap Epigenomics Consortium et al. Integrative analysis of 111 reference human epigenomes Nature 518, 317–330 (2015).

  64. Krzywinski, M. et al. Circos: an information aesthetic for comparative genomics. Genome Res. 19, 1639–1645 (2009).

    Article  CAS  Google Scholar 

  65. Zheng, Y. et al. m6AVar: a database of functional variants involved in m6A modification. Nucleic Acids Res. 46, D139–D145 (2018).

    Article  CAS  Google Scholar 

  66. MacArthur, J. et al. The new NHGRI-EBI Catalog of published genome-wide association studies (GWAS Catalog). Nucleic Acids Res. 45, D896–D901 (2017).

    Article  CAS  Google Scholar 

  67. Maurano, M. T. et al. Systematic localization of common disease-associated variation in regulatory DNA. Science 337, 1190–1195 (2012).

    Article  CAS  Google Scholar 

  68. Yu, G., Wang, L. G. & He, Q. Y. ChIPseeker: an R/Bioconductor package for ChIP peak annotation, comparison and visualization. Bioinformatics 31, 2382–2383 (2015).

    Article  CAS  Google Scholar 

  69. Saxonov, S., Berg, P. & Brutlag, D. L. A genome-wide analysis of CpG dinucleotides in the human genome distinguishes two distinct classes of promoters. Proc. Natl Acad. Sci. USA 103, 1412–1417 (2006).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We thank K. Yen for critical reading of the manuscript, and J. Chen and Z. Zuo for discussion about this project. This work was supported by the National Key R&D Program of China (2017YFA0106700 and 2018YFC1004103), the Natural Science Foundation of China (31722034, 81671466, 81870129 and 81771643), Innovation Team in Wisdom Valley of Southern China (2015CXDT06) and Pearl River S&T Nova Program of Guangzhou (201806010009).

Author information

Authors and Affiliations

Authors

Contributions

S.X. and Laixin X. conceived the research. M.Z., C.H. and Laixin X. designed and supervised the project. S.X., Q.H., X.L., J.S., W.W., Y.L., G.J., M.D., S.L., H.Z., M.Y., K.T. and Q.L. performed experiments. S.X., S.C. and Linjian X. conducted bioinformatics analysis. S.X. and Laixin X. wrote the manuscript with input from all authors.

Corresponding authors

Correspondence to Mei Zhong, Chunjiang He or Laixin Xia.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Publisher’s note: Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Integrated supplementary information

Supplementary Figure 1 Whole-transcriptome profiling profiling of m6As in major human tissues using an improved MeRIP procedure.

a. The non-specific rate (left) of MeRIP using normal or competitive washing. The non-specific IP rate was calculated from the LC/MS-MS data using the ratio of m(6)A/A in the input sample divided by that in the IP sample. Meanwhile, ssRNA with or without m(6)A was used to validated the signal to noise ratio of these two methods (right). Enrichment of IPed versus input RNA is normalize against ssRNA without m(6)A. Two independent experiments were performed with bar representing the average value (see source data in Supplementary Table 5). b. Similarity (Pearson’s correlation) of gene expression levels between each pair of samples. The FPKM of genes from input were used. The samples were hierarchically clustered (see source data in Supplementary Table 5). c. Fraction of overlapped m(6)A peaks called by MACS2 and MeTPeak/moving-window. d. Sequence logo and p value of deduced consensus motif of m(6)As in each tissue. n= 36461, 27866, 45503, 26163, 24180, 29174, 38444, 21780 m(6)As in brain, heart, kidney, liver, lung, muscle, placenta and stomach respectively, binomial distribution test (see methods for details). e. Distribution of m(6)A peaks surrounding the CDS in mRNA regions in each tissue. f. The fraction of m(6)A peaks in heart, kidney, liver, lung, muscle, placenta, and stomach in the 5′ UTR, CDS, intron, stop codon, 3′ UTR, promoter, and intergenic regions.

Supplementary Figure 2 m6As on intronic regions in human tissues.

a. MeRIP enrichment (upper) and expression level (lower) of m(6)A-negative (blue) and positive (orange) intronic regions in liver and stomach. Both the enrichment of IPed versus input RNA and the expression level are normalized against GAPDH. MeRIP-qPCR and qPCR were performed once with two technical replicates obtained and shown (see source data in Supplementary Table 5). b. Sequence logo and p value of deduced consensus motif of 81981 intronic m(6)As with strand information. Binomial distribution test (see methods for details). c. Ratio of uniquely mapped reads in intronic regions. Biologically independent samples n=3, 3, 3, 3, 2, 2, 3, 2 in brain, heart, kidney, liver, lung, muscle, placenta and stomach, respectively (see source data in Supplementary Table 5). d. Fraction of intronic m(6)As in each tissue determined in a rigorously way as described in methods. e. Histogram of completed splicing index (coSI) values for internal exon in each tissue.

Supplementary Figure 3 Differential m6As among human tissues.

a. Circos plot of tissue-differential m(6)As across the genome in brain (orange), heart (blue), kidney (green), liver (gray), lung (acen), muscle (purple), placenta (red), and stomach (yellow), respectively.b. MeRIP enrichment (upper) and expression level (lower) of two lung-differential m(6)A peaks on ECT2L and WNT4, and one stomach-differential peak on TMEM184A. Enrichment of IPed versus input RNA and the expression level is normalize against ZNF384, which is a conserved m(6)A peak across all tissues. MeRIP-qPCR and qPCR were performed once with two technical replicates obtained and shown.NA means the RNA level is too low to be detected by qPCR (see source data in Supplementary Table 5).

Supplementary Figure 4 m6As on lincRNA in human tissues.

a. MeRIP enrichment (upper) and expression level (lower) of m(6)A-negative and positive regions on e-lincRNA and other lincRNA in lung and stomach. Enrichment of IPed versus input RNA and the expression level is normalize against GAPDH. MeRIP-qPCR and qPCR were performed once with three technical replicates obtained and shown (see source data in Supplementary Table 5). b. Sequence logo and p value of deduced consensus motif of 226588 m(6)As with strand information resided in mRNA. Binomial distribution test. c. Number of mRNA modified by m(6)A or not in each tissue.

Supplementary Figure 5 eQTL and GWAS SNPs are associated with fetal tissue m6As.

a. Odds ratio of eQTL SNPs enriched in m(6)As across all HapMap SNPs in corresponding tissues. n = 682487.04, 978250.18, 699224.11, 676464.39, 765139.26, 765776.76, 1391369.53, 1392390.1, 1395465.56, 1397037.35, 1394019.17, 1393978.32, 1396518.69, 1399635.18, 1404775.28, 1400517.83, 1398627.95, 1395485.53, 1392218 bases from the top down, respectively, two-sided Fisher’s exact test, P < 0.05 (see source data in Supplementary Table 5). b. Distribution of eQTL SNPs located in m(6)As in heart, liver, lung, muscle, and stomach in the 5′ UTR, CDS, intron, stop codon, 3′ UTR, promoter, and intergenic regions. c. Representative eQTL SNP on gene WNT2B (left) and GWAS SNP on gene HNF1A-AS1 (right). Normalized reads densities of MeRIP-Seq and Input-Seq of different tissues are shown. Reads of Input-Seq and MeRIP-Seq were gray and colored, respectively. All biological replicates are shown. Levels were normalized by the number of reads in each sample. Range is shown at the right side of the ‘brain-1’ track. The MeRIP-Seq was performed two or three times for each tissue type as indicated, and all replicates were shown. d. Enrichment of IPed versus input RNA of SNP-associated m(6)A peaks was normalized to GAPDH. MeRIP-qPCR was performed once with three technical replicates obtained and shown (see source data in Supplementary Table 5).

Supplementary Figure 6 Genes enriched in m6As have CpG-rich promoters.

a. Western blots of FLAG or ß-ACTIN from SFB-METTL3 Hela cells before and after doxycycline induced expression. Unprocessed data is shown in Supplementary Figure 8. The western blot was performed twice with similar results, and a representative figure was shown. b. Enrichment of METTL3 occupancy in the promoters of m(6)A modified genes over unmodified genes. N = 19801, 18676, 18439, 18030, 17567, 17374, 17778 and 17303 genes in brain, heart, kidney, liver, lung, muscle, placenta and stomach, respectively. One-sided fisher’s exact test, P <2.2E-16 (see source data in Supplementary Table 5). c. Guitar plot of m(6)As distribution on genes with CpG-rich or poor promoters.

Supplementary Figure 7 The CpG status of promoters is involved in m6A methylation.

a. m(6)A distribution on mouse gene Ankrd9 and Zbtb42. Normalized reads densities of IP (orange) versus input (gray) were shown. b. RNA expression level of POU5F1/EEF1A1-Ankrd9/Zbtb42 in Hela cells. The expression level of Ankrd9 or Zbtb42 was first normalize to ACTB, and then relative to that with EEF1A1 promoters. Error bars represent mean s.d. from three independent experiments (see source data in Supplementary Table 5). c. Protein level of Ankrd9/Zbtb42, ACTB, IRF1, SMAD7 and GAPDH in promoter-displaced Hela cells. MYC tag placed before Ankrd9 or Zbtb42 coding regions was used to determine the protein level of them. Unprocessed data is shown in Supplementary Figure 8. The western blot was performed twice with similar results, and a representative figure was shown. d. Expression level of POU5F1/EEF1A1-Ankrd9/Zbtb42 in HEK293 cells. The relative expression level of Ankrd9 or Zbtb42 was judged as aforementioned. Two independent experiments were shown (see source data in Supplementary Table 5). e. Protein level of Ankrd9/Zbtb42, ACTB, IRF1 and GAPDH in promoter-displaced HEK293 cells. Unprocessed data is shown in Supplementary Figure 8. The western blot was performed twice with similar results, and a representative figure was shown. f. The relative m6A levels of ACTB, Ankrd9 and Zbtb42 in promoter displaced HEK293 cells were firstly normalized to their own inputs and then relative to GAPDH in corresponding cells. The experiments were performed twice (see source data in Supplementary Table 5). g. Lifetime of Ankrd9, Zbtb42, and IRF1 in promoter-displaced HEK293 cells after actinomycin D treatment. The plotted points represented the individual data from two independent experiments (see source data in Supplementary Table 5). h. Depth of CEBPZ and SMAD2 ChIP-Seq fragments (per bp per peak) around CpG-rich or CpG-poor TSS regions.

Supplementary Figure 8 Uncropped images from Western blots.

Uncropped images for Supplementary Fig.6a, 7c and 7e.

Supplementary information

Supplementary Information

Supplementary Figures 1–8 and legends for Supplementary Tables 1–5

Reporting Summary

Supplementary Table 1

Sample information in this study.

Supplementary Table 2

A summary of MeRIP sequencing statistics for each sample.

Supplementary Table 3

Cis-eQTLs SNPs in fetal tissue m6A regions potentially affecting m6A sites in corresponding tissues.

Supplementary Table 4

List of primers used in this study.

Supplementary Table 5

Statistics source data.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Xiao, S., Cao, S., Huang, Q. et al. The RNA N6-methyladenosine modification landscape of human fetal tissues. Nat Cell Biol 21, 651–661 (2019). https://doi.org/10.1038/s41556-019-0315-4

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41556-019-0315-4

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