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:

mEnrich-seq: methylation-guided enrichment sequencing of bacterial taxa of interest from microbiome

Abstract

Metagenomics has enabled the comprehensive study of microbiomes. However, many applications would benefit from a method that sequences specific bacterial taxa of interest, but not most background taxa. We developed mEnrich-seq (in which ‘m’ stands for methylation and seq for sequencing) for enriching taxa of interest from metagenomic DNA before sequencing. The core idea is to exploit the self versus nonself differentiation by natural bacterial DNA methylation and rationally choose methylation-sensitive restriction enzymes, individually or in combination, to deplete host and background taxa while enriching targeted taxa. This idea is integrated with library preparation procedures and applied in several applications to enrich (up to 117-fold) pathogenic or beneficial bacteria from human urine and fecal samples, including species that are hard to culture or of low abundance. We assessed 4,601 bacterial strains with mapped methylomes so far and showed broad applicability of mEnrich-seq. mEnrich-seq provides microbiome researchers with a versatile and cost-effective approach for selective sequencing of diverse taxa of interest.

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: mEnrich-seq workflow and its application.
Fig. 2: Validation of mEnrich-seq.
Fig. 3: Enrichment of A. muciniphila from fecal samples by mEnrich-seq.
Fig. 4: Enrichment sequencing of bacterial taxa based on de novo discovered methylation motifs.
Fig. 5: Assessing the broad applicability of mEnrich-seq.

Similar content being viewed by others

Data availability

All sequencing data generated for this study have been uploaded to the Sequence Read Archive under the BioProject PRJNA896537. Source data are provided with this paper.

Code availability

Primary sequence data analysis was done using open-source software tools as described in the Methods. Custom scripts used to analyze motif frequency are available in Zenodo at https://doi.org/10.5281/zenodo.8330164 (ref. 71).

References

  1. Lynch, S. V. & Pedersen, O. The human intestinal microbiome in health and disease. N. Engl. J. Med. 375, 2369–2379 (2016).

    Article  CAS  PubMed  Google Scholar 

  2. Cho, I. & Blaser, M. J. The human microbiome: at the interface of health and disease. Nat. Rev. Genet. 13, 260–270 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Pryor, R., Martinez-Martinez, D., Quintaneiro, L. & Cabreiro, F. The role of the microbiome in drug response. Annu. Rev. Pharmacol. Toxicol. 60, 417–435 (2020).

    Article  CAS  PubMed  Google Scholar 

  4. Hedin, C. R. et al. Altered intestinal microbiota and blood T cell phenotype are shared by patients with Crohn’s disease and their unaffected siblings. Gut 63, 1578–1586 (2014).

    Article  CAS  PubMed  Google Scholar 

  5. Qin, J. et al. A metagenome-wide association study of gut microbiota in type 2 diabetes. Nature 490, 55–60 (2012).

    Article  CAS  PubMed  ADS  Google Scholar 

  6. Joossens, M. et al. Dysbiosis of the faecal microbiota in patients with Crohn’s disease and their unaffected relatives. Gut 60, 631–637 (2011).

    Article  PubMed  Google Scholar 

  7. Hsiao, E. Y. et al. Microbiota modulate behavioral and physiological abnormalities associated with neurodevelopmental disorders. Cell 155, 1451–1463 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Derrien, M., Belzer, C. & de Vos, W. M. Akkermansia muciniphila and its role in regulating host functions. Micro. Pathog. 106, 171–181 (2017).

    Article  Google Scholar 

  9. Lugli, G. A. et al. Isolation of novel gut bifidobacteria using a combination of metagenomic and cultivation approaches. Genome Biol. 20, 96 (2019).

  10. Chiu, C. Y. & Miller, S. A. Clinical metagenomics. Nat. Rev. Genet. 20, 341–355 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Loman, N. J. et al. A culture-independent sequence-based metagenomics approach to the investigation of an outbreak of Shiga-toxigenic Escherichia coli O104: H4. J. Am. Med. Assoc. 309, 1502–1510 (2013).

    Article  CAS  ADS  Google Scholar 

  12. Johns, N. I. et al. Metagenomic mining of regulatory elements enables programmable species-selective gene expression. Nat. Methods 15, 323–329 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Greub, G. Culturomics: a new approach to study the human microbiome. Clin. Microbiol. Infect. 18, 1157–1159 (2012).

    Article  CAS  PubMed  Google Scholar 

  14. Lagier, J.-C. et al. Microbial culturomics: paradigm shift in the human gut microbiome study. Clin. Microbiol. Infect. 18, 1185–1193 (2012).

    Article  CAS  PubMed  Google Scholar 

  15. Meyer, F. et al. Critical assessment of metagenome interpretation: the second round of challenges. Nat. Methods 19, 429–440 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Laudadio, I., Fulci, V., Stronati, L. & Carissimi, C. Next-generation metagenomics: methodological challenges and opportunities. OMICS 23, 327–333 (2019).

    Article  CAS  PubMed  Google Scholar 

  17. Simon, H. Y., Siddle, K. J., Park, D. J. & Sabeti, P. C. Benchmarking metagenomics tools for taxonomic classification. Cell 178, 779–794 (2019).

    Article  Google Scholar 

  18. Cleary, B. et al. Detection of low-abundance bacterial strains in metagenomic datasets by eigengenome partitioning. Nat. Biotechnol. 33, 1053–1060 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Martin, S. et al. Nanopore adaptive sampling: a tool for enrichment of low abundance species in metagenomic samples. Genome Biol. 23, 11 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Charalampous, T. et al. Nanopore metagenomics enables rapid clinical diagnosis of bacterial lower respiratory infection. Nat. Biotechnol. 37, 783–792 (2019).

    Article  CAS  PubMed  Google Scholar 

  21. Hasan, M. R. et al. Depletion of human DNA in spiked clinical specimens for improvement of sensitivity of pathogen detection by next-generation sequencing. J. Clin. Microbiol. 54, 919–927 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Feehery, G. R. et al. A method for selectively enriching microbial DNA from contaminating vertebrate host DNA. PLoS ONE 8, e76096 (2013).

    Article  CAS  PubMed  PubMed Central  ADS  Google Scholar 

  23. Ji, P., Zhang, Y., Wang, J. & Zhao, F. MetaSort untangles metagenome assembly by reducing microbial community complexity. Nat. Commun. 8, 14306 (2017).

    Article  CAS  PubMed  PubMed Central  ADS  Google Scholar 

  24. Loose, M., Malla, S. & Stout, M. Real-time selective sequencing using nanopore technology. Nat. Methods 13, 751–754 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Kovaka, S., Fan, Y., Ni, B., Timp, W. & Schatz, M. C. Targeted nanopore sequencing by real-time mapping of raw electrical signal with UNCALLED. Nat. Biotechnol. 39, 431–441 (2021).

    Article  CAS  PubMed  Google Scholar 

  26. Payne, A. et al. Readfish enables targeted nanopore sequencing of gigabase-sized genomes. Nat. Biotechnol. 39, 442–450 (2021).

    Article  CAS  PubMed  Google Scholar 

  27. Liu, J. et al. The fecal resistome of dairy cattle is associated with diet during nursing. Nat. Commun. 10, 4406 (2019).

    Article  PubMed  PubMed Central  ADS  Google Scholar 

  28. Aggarwala, V. et al. Precise quantification of bacterial strains after fecal microbiota transplantation delineates long-term engraftment and explains outcomes. Nat. Microbiol. 6, 1309–1318 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Sánchez-Romero, M. A. & Casadesús, J. The bacterial epigenome. Nat. Rev. Microbiol. 18, 7–20 (2020).

    Article  PubMed  Google Scholar 

  30. Casadesús, J. & Low, D. Epigenetic gene regulation in the bacterial world. Microbiol. Mol. Biol. Rev. 70, 830–856 (2006).

    Article  PubMed  PubMed Central  Google Scholar 

  31. Beaulaurier, J., Schadt, E. E. & Fang, G. Deciphering bacterial epigenomes using modern sequencing technologies. Nat. Rev. Genet. 20, 157–172 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Beaulaurier, J. et al. Single molecule-level detection and long read-based phasing of epigenetic variations in bacterial methylomes. Nat. Commun. 6, 7438 (2015).

    Article  CAS  PubMed  ADS  Google Scholar 

  33. Blow, M. J. et al. The epigenomic landscape of prokaryotes. PLoS Genet. 12, e1005854 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  34. Tourancheau, A., Mead, E. A., Zhang, X.-S. & Fang, G. Discovering multiple types of DNA methylation from bacteria and microbiome using nanopore sequencing. Nat. Methods 18, 491–498 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Beaulaurier, J. et al. Metagenomic binning and association of plasmids with bacterial host genomes using DNA methylation. Nat. Biotechnol. 36, 61–69 (2018).

    Article  CAS  PubMed  Google Scholar 

  36. Conlan, S. et al. Single-molecule sequencing to track plasmid diversity of hospital-associated carbapenemase-producing Enterobacteriaceae. Sci. Transl. Med. 6, 254ra126 (2014).

  37. Kobayashi, I., Nobusato, A., Kobayashi-Takahashi, N. & Uchiyama, I. Shaping the genome–restriction–modification systems as mobile genetic elements. Curr. Opin. Genet. Dev. 9, 649–656 (1999).

    Article  CAS  PubMed  Google Scholar 

  38. Enam, S. U. et al. Restriction endonuclease-based modification-dependent enrichment (REMoDE) of DNA for metagenomic sequencing. Appl. Environ. Microbiol. 89, e0167022 (2023).

  39. Oliveira, P. H. & Fang, G. Conserved DNA methyltransferases: a window into fundamental mechanisms of epigenetic regulation in bacteria. Trends Microbiol. 29, 28–40 (2021).

    Article  CAS  PubMed  Google Scholar 

  40. Fang, G. et al. Genome-wide mapping of methylated adenine residues in pathogenic Escherichia coli using single-molecule real-time sequencing. Nat. Biotechnol. 30, 1232–1239 (2012).

    Article  CAS  PubMed  Google Scholar 

  41. Kong, Y. et al. Critical assessment of DNA adenine methylation in eukaryotes using quantitative deconvolution. Science 375, 1979 (2022).

    Article  Google Scholar 

  42. Mirkin, S. M. Expandable DNA repeats and human disease. Nature 447, 932–940 (2007).

    Article  CAS  PubMed  ADS  Google Scholar 

  43. Wang, G. & Vasquez, K. M. Dynamic alternative DNA structures in biology and disease. Nat. Rev. Genet. 24, 211–234 (2023).

  44. Urinary Tract Infection (Catheter-Associated Urinary Tract Infection (CAUTI) and Non-Catheter-Associated Urinary Tract Infection (UTI)) Events (National Healthcare Safety Network, 2022).

  45. Cronquist, A. B. et al. Impacts of culture-independent diagnostic practices on public health surveillance for bacterial enteric pathogens. Clin. Infect. Dis. 54, S432–S439 (2012).

    Article  PubMed  Google Scholar 

  46. Moustafa, A. et al. Microbial metagenome of urinary tract infection. Sci. Rep. 8, 4333 (2018).

    Article  PubMed  PubMed Central  ADS  Google Scholar 

  47. Derrien, M., Collado, M. C., Ben-Amor, K., Salminen, S. & de Vos, W. M. The Mucin degrader Akkermansia muciniphila is an abundant resident of the human intestinal tract. Appl. Environ. Microbiol. 74, 1646–1648 (2008).

    Article  CAS  PubMed  ADS  Google Scholar 

  48. Collado, M. C., Derrien, M., Isolauri, E., de Vos, W. M. & Salminen, S. Intestinal integrity and Akkermansia muciniphila, a mucin-degrading member of the intestinal microbiota present in infants, adults, and the elderly. Appl. Environ. Microbiol. 73, 7767–7770 (2007).

    Article  CAS  PubMed  PubMed Central  ADS  Google Scholar 

  49. Dao, M. C. et al. Akkermansia muciniphila and improved metabolic health during a dietary intervention in obesity: relationship with gut microbiome richness and ecology. Gut 65, 426–436 (2016).

    Article  CAS  PubMed  Google Scholar 

  50. Everard, A. et al. Cross-talk between Akkermansia muciniphila and intestinal epithelium controls diet-induced obesity. Proc. Natl Acad. Sci. USA 110, 9066–9071 (2013).

    Article  CAS  PubMed  PubMed Central  ADS  Google Scholar 

  51. Lee, K. A. et al. Cross-cohort gut microbiome associations with immune checkpoint inhibitor response in advanced melanoma. Nat. Med. 28, 535–544 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Routy, B. et al. Gut microbiome influences efficacy of PD-1–based immunotherapy against epithelial tumors. Science 359, 91–97 (2018).

    Article  CAS  PubMed  ADS  Google Scholar 

  53. Derosa, L. et al. Intestinal Akkermansia muciniphila predicts clinical response to PD-1 blockade in patients with advanced non-small-cell lung cancer. Nat. Med. 28, 315–324 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Caputo, A. et al. Whole-genome assembly of Akkermansia muciniphila sequenced directly from human stool. Biol. Direct 10, 5 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  55. Bokulich, N. A. et al. Antibiotics, birth mode, and diet shape microbiome maturation during early life. Sci. Transl. Med. 8, 343ra82 (2016).

  56. Kolmogorov, M. et al. metaFlye: scalable long-read metagenome assembly using repeat graphs. Nat. Methods 17, 1103–1110 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Roberts, R. J., Vincze, T., Posfai, J. & Macelis, D. REBASE—a database for DNA restriction and modification: enzymes, genes and genomes. Nucleic Acids Res. 43, D298–D299 (2015).

    Article  CAS  PubMed  Google Scholar 

  58. Maghini, D. G., Moss, E. L., Vance, S. E. & Bhatt, A. S. Improved high-molecular-weight DNA extraction, nanopore sequencing and metagenomic assembly from the human gut microbiome. Nat. Protoc. 16, 458–471 (2021).

    Article  CAS  PubMed  Google Scholar 

  59. Gupta, R., Xu, S.-Y., Sharma, P. & Capalash, N. Characterization of MspNI (G/GWCC) and MspNII (R/GATCY), novel thermostable Type II restriction endonucleases from Meiothermus sp., isoschizomers of AvaII and BstYI. Mol. Biol. Rep. 39, 5607–5614 (2012).

    Article  CAS  PubMed  Google Scholar 

  60. Wu-Woods, N. J. et al. Microbial-enrichment method enables high-throughput metagenomic characterization from host-rich samples. Nat. Methods 20, 1672–1682 (2023).

    Article  CAS  PubMed  Google Scholar 

  61. Becken, B. et al. Genotypic and phenotypic diversity among human isolates of Akkermansia muciniphila. mBio 12, e00478–21 (2021).

  62. Cao, L et al. mEnrich-seq: methylation-guided enrichment sequencing of bacterial taxa of interest from microbiome, PROTOCOL (version 1). Protocol Exchange https://doi.org/10.21203/rs.3.pex-2373/v1 (2023).

  63. Optional fragmentation of gDNA. Oxford Nanopore Technology https://community.nanoporetech.com/extraction_method_groups/optional-fragmentation-of-gdna (2022).

  64. Bag, S., Ghosh, T. S. & Das, B. Complete genome sequence of Collinsella aerofaciens isolated from the gut of a healthy Indian subject. Genome Announc. 5, e01361−17 (2017).

  65. Buttimer, C. et al. Selective isolation of Eggerthella lenta from human faeces and characterisation of the species prophage diversity. Microorganisms 10, 195 (2022).

  66. Ribeiro, F. J. et al. Finished bacterial genomes from shotgun sequence data. Genome Res. 22, 2270–2277 (2012).

  67. Fukuda, S. et al. Bifidobacteria can protect from enteropathogenic infection through production of acetate. Nature 469, 543–547 (2011).

  68. O'Callaghan, A. & van Sinderen, D. Bifidobacteria and their role as members of the human gut microbiota. Front Microbiol. 7, 925 (2016).

  69. Uritskiy, G. V., DiRuggiero, J. & Taylor, J. MetaWRAP—a flexible pipeline for genome-resolved metagenomic data analysis. Microbiome 6, 158 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  70. Asnicar, F. et al. Precise phylogenetic analysis of microbial isolates and genomes from metagenomes using PhyloPhlAn 3.0. Nat. Commun. 11, 2500 (2020).

    Article  CAS  PubMed  PubMed Central  ADS  Google Scholar 

  71. Cao, L., Kong, Y. & Fang, G. Custom code for Nature Methods manuscript NMETH-A50860. Zenodo https://doi.org/10.5281/zenodo.8330164 (2023).

Download references

Acknowledgements

We thank the Icahn School of Medicine at Mount Sinai colleagues J. Faith for helping with the characterization of the adult fecal sample and F. Samaroo for helping with urine sample processing; L. Davey and R. Valdivia at Duke University School of Medicine for help with isolation of A. muciniphila; and the M. Blaser laboratory at the Rutgers University and other research teams that collected the infant fecal samples for The Early Childhood Antibiotics and the Microbiome study. This work was supported by grant no. R35 GM139655 (G.F.) from the National Institutes of Health. G.F. is a Hirschl Research Scholar by Irma T. Hirschl/Monique Weill-Caulier Trust and a Nash Family Research Scholar. This work was also supported in part through the computational resources and staff expertise provided by the Department of Scientific Computing at the Icahn School of Medicine at Mount Sinai.

Author information

Authors and Affiliations

Authors

Contributions

G.F. conceived and supervised the project. L.C. and G.F. designed the methods. L.C. performed all mEnrich-seq experiments and sequencing. Y.K., Y.F., M.N. and A.T. performed the evaluation of mEnrich-seq and analyzed mEnrich-seq data across the applications. M.K. helped with the data analysis of mEnrich-seq of the urine samples. T.K. and M.G. helped with the characterization of urine samples and E. coli strains. X.-S.Z. helped with the characterization of the infant fecal samples. L.C., M.N. and E.A.M. performed additional sequencing. L.C., Y.F., M.N., X.-S.Z. and G.F. performed additional data analyses. L.C. and G.F. wrote the manuscript with inputs and comments from all coauthors.

Corresponding author

Correspondence to Gang Fang.

Ethics declarations

Competing interests

L.C. and G.F. are the coinventors of a US provisional patent application (application no.: 63/382,616) based on the method described in this work. The other authors declare no competing interests.

Peer review

Peer review information

Nature Methods thanks the anonymous reviewers for their contribution to the peer review of this work. Primary Handling Editor: Lin Tang, in collaboration with the Nature Methods team.

Additional information

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

Extended data

Extended Data Fig. 1 mEnrich-seq of urine samples using Nanopore Flongle flow cells.

a, Three urine samples were sequenced as the input: UTI-1 contained mostly bacterial DNA, while UTI-2 and UTI-3 were dominated by human DNA. b, Scatter plots for six urine samples (UTI-1 to -6) showing the percentage of reads mapped to E. coli, human and other taxa from the input (x-axis) vs. mEnrich-seq (y-axis). The UTI-1 to -3 samples demonstrate consistent patterns as shown in Fig. 2 in the main text (MinION flow cell); the additional three UTI urine samples (UTI -4 to -6) are included to illustrate the consistent enrichment of E. coli by mEnrich-seq. Across all six samples, E. coli reads are significantly enriched in mEnrich-seq. In UTI-2, UTI-3, UTI-4, and UTI-6, the proportion of reads corresponding to the human genome are significantly lower due to cleavage by DpnII in mEnrich-seq. c, Fold enrichment of E. coli reads from mEnrich-seq compared to the input for the six urine samples.

Source data

Extended Data Fig. 2 mEnrich-seq data of UTI-1 include reads that have high-confidence mapping to E. coli yet carry additional genes beyond the isolated strain from standard urine culture.

a, Top: 10 reads with highly confident mapping to E. coli from mEnrich-seq of UTI-1 urine sample, consistently supporting a tet(A) resistant gene (from AF534183, blue boxes). For each read, lengths of the sequences flanking the resistance gene were shown on both sides. The identities between the reads and tet(A) were shown inside the blue boxes. Bottom, reads alignment to tet(A) was performed with Clustalw2 and colored with MView. b, Validation of tet(A) in UTI-1 sample. Agarose gel electrophoresis of the tet(A) regions amplified once by two sets of primers (of lengths 1150 bp and 900 bp, respectively). The bands (in red square) were further purified for gene sequence confirmation by Sanger sequencing (Supplementary Information).

Source data

Extended Data Fig. 3 Confirmation of DNA 6 mA methylation at RA6mATTY motif sites using PacBio sequencing of two A. muciniphila strains.

a, RA6mATTY (mediated by MTase AmuORF1905P) is one of the five methylated motifs detected in A. muciniphila (ATCC BAA-835) by PacBio sequencing as summarized in REBASE. b, DNA 6 mA methylation at RAATTY motif sites (light blue) is confirmed based on the high IPD ratios observed in PacBio sequencing data of the A. muciniphila strain isolated from the infant fecal sample GUT-3. c, DNA 6mA methylation at RAATTY motif sites (purple) is confirmed based on the high IPD ratio observed in PacBio SMRT sequencing data of another A. muciniphila strain.

Extended Data Fig. 4 De novo methylation motif discovery from an initial shallow sequencing run (400k PacBio Sequel II CCS reads).

Many methylation motifs were discovered from SMRT sequencing data, even those from bacterial genomes with ~10% completeness. The relative abundance of each bacterial genome is included in the x-axis label in red color. 4-mer motifs (those with four non-degenerate bases) and similarly 5-mer and 6-mer motifs are color coded.

Source data

Extended Data Fig. 5 Direct comparison shows that the mEnrich-seq design has better fold enrichment than REMoDE.

a, Agarose gel electrophoresis of Mock-3 (a mock with five species, see Supplementary Information) after treatment by RE and T5 exonuclease for once. Left, Agarose gel electrophoresis of the mock sample digested by RE, XapI. Due to the sparse RE recognition sites in the background genomes in the mock microbiome sample, relatively long DNA fragments from the background genomes remain even after RE digestion; Right, The gel image of the mock sample digested by RE and T5. b, A direct comparison between mEnrich-seq and REMoDE was performed using the same mock microbiome sample (Mock-3), and the same sequencing platform (Illumina). To be compatible with the short-read sequencing platform, the library of mEnrich-seq was prepared according to the ‘Compatibility between mEnrich-seq and other sequencing platforms’ section in the Supplementary Information.

Source data

Extended Data Fig. 6 mEnrich-seq design has better fold enrichment than REMoDE when they are compared with the same, real microbiome samples with DNA fragmentation and damages.

a, Agarose gel electrophoresis tested once on genomic DNA isolated from Mock-3 (illustrating the ideal high molecular weight gDNA from cell culture) and two infant fecal samples (representing DNA quality in real applications). b, Comparing the fold enrichment of A. muciniphila from two infant fecal samples using mEnrich-seq, REMoDE, or REMoDE with DNA repair beforehand (see Supplementary Information). As DNA repair is part of mEnrich-seq by default, this step is also applied to REMoDE for a fair comparison.

Source data

Extended Data Fig. 7 Evaluation of the benefit of adapter ligation before vs. after RE-digestion using the Mock-3.

The bar graph demonstrates that adapter ligation before RE-digestion indeed has better fold enrichment (see Methods in Supplementary Information).

Source data

Extended Data Fig. 8 An alternative adapter has comparable enrichment efficiency as the original adapter.

The fold enrichment of A. muciniphila (using XapI as RE) was tested on Mock-3 with either the original adapter or the alternative adapter (sequences in Supplementary Table 9).

Source data

Extended Data Fig. 9 GC content does not introduce systematic bias on read coverage across the E. coli genome analyzed by mEnrich-seq.

Analysis was performed on Mock-1 and Mock-2 (E. coli relative abundance as 0.91% and 1.20%, see Supplementary Information). For a fair comparison, the same yield of data from both input and mEnrich-seq were used in data analysis and figures. Each dot represents a 5 kb genomic region, colored by GC content. X-axis: number of input reads mapped to each 5 kb genomic region; Y-axis: number of mEnrich-seq reads mapped to each 5 kb genomic region. To ease visualization, a random value was added on x-values to avoid overlapping dots.

Source data

Extended Data Fig. 10 GC content does not introduce systematic bias on read coverage across the A. muciniphila genome by mEnrich-seq.

The same type of scatter plots (as the ones above for E. coli) made with sequencing data from three human gut samples (A. muciniphila relative abundance: 0.48%, 0.52% and 1.52%, respectively) to characterize the enrichment of A. muciniphila. Consistent with the Circos plot for the same samples, read depths were capped at the 95% quantile to ease visualization.

Source data

Supplementary information

Supplementary Information

Supplementary text, Figs. 1–4, protocols and unprocessed gels for Figs. 3 and 4.

Reporting Summary

Supplementary Tables

Supplementary Tables 1–13.

Source data

Source Data Fig. 2

Statistical source data.

Source Data Fig. 3

Statistical source data.

Source Data Fig. 4

Statistical source data.

Source Data Fig. 5

Statistical source data.

Source Data Extended Data Figs. 1 and 4–10

Statistical source data.

Source Data Extended Data Fig. 2

Unprocessed gel.

Source Data Extended Data Fig. 5

Unprocessed gel.

Source Data Extended Data Fig. 6

Unprocessed gel.

Rights and permissions

Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Cao, L., Kong, Y., Fan, Y. et al. mEnrich-seq: methylation-guided enrichment sequencing of bacterial taxa of interest from microbiome. Nat Methods 21, 236–246 (2024). https://doi.org/10.1038/s41592-023-02125-1

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41592-023-02125-1

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