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.

  • Analysis
  • Published:

Measurement of bacterial replication rates in microbial communities

Abstract

Culture-independent microbiome studies have increased our understanding of the complexity and metabolic potential of microbial communities. However, to understand the contribution of individual microbiome members to community functions, it is important to determine which bacteria are actively replicating. We developed an algorithm, iRep, that uses draft-quality genome sequences and single time-point metagenome sequencing to infer microbial population replication rates. The algorithm calculates an index of replication (iRep) based on the sequencing coverage trend that results from bi-directional genome replication from a single origin of replication. We apply this method to show that microbial replication rates increase after antibiotic administration in human infants. We also show that uncultivated, groundwater-associated, Candidate Phyla Radiation bacteria only rarely replicate quickly in subsurface communities undergoing substantial changes in geochemistry. Our method can be applied to any genome-resolved microbiome study to track organism responses to varying conditions, identify actively growing populations and measure replication rates for use in modeling studies.

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: iRep determines replication rates for bacteria using genome-resolved metagenomics.
Figure 2: iRep is an accurate measure of in situ replication rates.
Figure 3: iRep and bPTR calculations agree for a novel Deltaproteobacterium sampled from groundwater.
Figure 4: Replication rates were determined for CPR and human microbiome-associated organisms.
Figure 5: Elevated replication rates are associated with antibiotic administration and were detected before onset of necrotizing enterocolitis (NEC) in premature infants.
Figure 6: Absolute abundance (bars, left axis) and iRep (scatter plot, right axis) values for bacterial species associated with two premature infants.

Similar content being viewed by others

Accession codes

Primary accessions

BioProject

Sequence Read Archive

References

  1. Bremer, H. & Churchward, G. An examination of the Cooper-Helmstetter theory of DNA replication in bacteria and its underlying assumptions. J. Theor. Biol. 69, 645–654 (1977).

    Article  CAS  Google Scholar 

  2. Skovgaard, O., Bak, M., Løbner-Olesen, A. & Tommerup, N. Genome-wide detection of chromosomal rearrangements, indels, and mutations in circular chromosomes by short read sequencing. Genome Res. 21, 1388–1393 (2011).

    Article  CAS  Google Scholar 

  3. Prescott, D.M. & Kuempel, P.L. Bidirectional replication of the chromosome in Escherichia coli. Proc. Natl. Acad. Sci. USA 69, 2842–2845 (1972).

    Article  CAS  Google Scholar 

  4. Wake, R.G. Visualization of reinitiated chromosomes in Bacillus subtilis. J. Mol. Biol. 68, 501–509 (1972).

    Article  CAS  Google Scholar 

  5. Sernova, N.V. & Gelfand, M.S. Identification of replication origins in prokaryotic genomes. Brief. Bioinform. 9, 376–391 (2008).

    Article  CAS  Google Scholar 

  6. Gao, F., Luo, H. & Zhang, C.-T. DoriC 5.0: an updated database of oriC regions in both bacterial and archaeal genomes. Nucleic Acids Res. 41, D90–D93 (2013).

    Article  CAS  Google Scholar 

  7. Anantharaman, K. et al. Analysis of five complete genome sequences for members of the class Peribacteria in the recently recognized Peregrinibacteria bacterial phylum. PeerJ 4, e1607 (2016).

    Article  Google Scholar 

  8. Korem, T. et al. Growth dynamics of gut microbiota in health and disease inferred from single metagenomic samples. Science 349, 1101–1106 (2015).

    Article  CAS  Google Scholar 

  9. Cooper, S. & Helmstetter, C.E. Chromosome replication and the division cycle of Escherichia coli B/r. J. Mol. Biol. 31, 519–540 (1968).

    Article  CAS  Google Scholar 

  10. Tyson, G.W. et al. Community structure and metabolism through reconstruction of microbial genomes from the environment. Nature 428, 37–43 (2004).

    Article  CAS  Google Scholar 

  11. Baker, B.J. et al. Enigmatic, ultrasmall, uncultivated Archaea. Proc. Natl. Acad. Sci. USA 107, 8806–8811 (2010).

    Article  CAS  Google Scholar 

  12. Sharon, I. et al. Time series community genomics analysis reveals rapid shifts in bacterial species, strains, and phage during infant gut colonization. Genome Res. 23, 111–120 (2013).

    Article  CAS  Google Scholar 

  13. Iverson, V. et al. Untangling genomes from metagenomes: revealing an uncultured class of marine Euryarchaeota. Science 335, 587–590 (2012).

    Article  CAS  Google Scholar 

  14. Nielsen, H.B. et al. Identification and assembly of genomes and genetic elements in complex metagenomic samples without using reference genomes. Nat. Biotechnol. 32, 822–828 (2014).

    Article  CAS  Google Scholar 

  15. Brown, C.T. et al. Unusual biology across a group comprising more than 15% of domain Bacteria. Nature 523, 208–211 (2015).

    Article  CAS  Google Scholar 

  16. Castelle, C.J. et al. Genomic expansion of domain archaea highlights roles for organisms from new phyla in anaerobic carbon cycling. Curr. Biol. 25, 690–701 (2015).

    Article  CAS  Google Scholar 

  17. Seitz, K.W., Lazar, C.S., Hinrichs, K.-U., Teske, A.P. & Baker, B.J. Genomic reconstruction of a novel, deeply branched sediment archaeal phylum with pathways for acetogenesis and sulfur reduction. ISME J. 10, 1696–1705 (2016).

    Article  CAS  Google Scholar 

  18. Joshi, N. Sickle. github.com https://github.com/najoshi/sickle.

  19. Peng, Y., Leung, H.C.M., Yiu, S.M. & Chin, F.Y.L. IDBA-UD: a de novo assembler for single-cell and metagenomic sequencing data with highly uneven depth. Bioinformatics 28, 1420–1428 (2012).

    Article  CAS  Google Scholar 

  20. Dick, G.J. et al. Community-wide analysis of microbial genome sequence signatures. Genome Biol. 10, R85 (2009).

    Article  Google Scholar 

  21. Wu, Y.-W., Simmons, B.A. & Singer, S.W. MaxBin 2.0: an automated binning algorithm to recover genomes from multiple metagenomic datasets. Bioinformatics 32, 605–607 (2016).

    Article  CAS  Google Scholar 

  22. Alneberg, J. et al. Binning metagenomic contigs by coverage and composition. Nat. Methods 11, 1144–1146 (2014).

    Article  CAS  Google Scholar 

  23. Parks, D.H., Imelfort, M., Skennerton, C.T., Hugenholtz, P. & Tyson, G.W. CheckM: assessing the quality of microbial genomes recovered from isolates, single cells, and metagenomes. Genome Res. 25, 1043–1055 (2015).

    Article  CAS  Google Scholar 

  24. Wrighton, K.C. et al. Fermentation, hydrogen, and sulfur metabolism in multiple uncultivated bacterial phyla. Science 337, 1661–1665 (2012).

    Article  CAS  Google Scholar 

  25. Di Rienzi, S.C. et al. The human gut and groundwater harbor non-photosynthetic bacteria belonging to a new candidate phylum sibling to Cyanobacteria. eLife 2, e01102 (2013).

    Article  Google Scholar 

  26. Castelle, C.J. et al. Extraordinary phylogenetic diversity and metabolic versatility in aquifer sediment. Nat. Commun. 4, 2120 (2013).

    Article  Google Scholar 

  27. Eloe-Fadrosh, E.A. et al. Global metagenomic survey reveals a new bacterial candidate phylum in geothermal springs. Nat. Commun. 7, 10476 (2016).

    Article  CAS  Google Scholar 

  28. Lobry, J.R. Asymmetric substitution patterns in the two DNA strands of bacteria. Mol. Biol. Evol. 13, 660–665 (1996).

    Article  CAS  Google Scholar 

  29. Raveh-Sadka, T. et al. Gut bacteria are rarely shared by co-hospitalized premature infants, regardless of necrotizing enterocolitis development. eLife 4, e05477 (2015).

    Article  Google Scholar 

  30. Sharon, I. et al. Accurate, multi-kb reads resolve complex populations and detect rare microorganisms. Genome Res. 25, 534–543 (2015).

    Article  CAS  Google Scholar 

  31. Paczia, N. et al. Extensive exometabolome analysis reveals extended overflow metabolism in various microorganisms. Microb. Cell Fact. 11, 122 (2012).

    Article  CAS  Google Scholar 

  32. Kopf, S.H. et al. Trace incorporation of heavy water reveals slow and heterogeneous pathogen growth rates in cystic fibrosis sputum. Proc. Natl. Acad. Sci. USA 113, E110–E116 (2016).

    Article  CAS  Google Scholar 

  33. Luef, B. et al. Diverse uncultivated ultra-small bacterial cells in groundwater. Nat. Commun. 6, 6372 (2015).

    Article  CAS  Google Scholar 

  34. Hug, L.A. et al. A new view of the tree of life. Nat. Microbiol. 1, 16048 (2016).

    Article  CAS  Google Scholar 

  35. Podar, M. et al. Targeted access to the genomes of low-abundance organisms in complex microbial communities. Appl. Environ. Microbiol. 73, 3205–3214 (2007).

    Article  CAS  Google Scholar 

  36. Rinke, C. et al. Insights into the phylogeny and coding potential of microbial dark matter. Nature 499, 431–437 (2013).

    Article  CAS  Google Scholar 

  37. Albertsen, M. et al. Genome sequences of rare, uncultured bacteria obtained by differential coverage binning of multiple metagenomes. Nat. Biotechnol. 31, 533–538 (2013).

    Article  CAS  Google Scholar 

  38. Kantor, R.S. et al. Small genomes and sparse metabolisms of sediment-associated bacteria from four candidate phyla. MBio 4, e00708–e00713 (2013).

    Article  Google Scholar 

  39. Nelson, W.C. & Stegen, J.C. The reduced genomes of Parcubacteria (OD1) contain signatures of a symbiotic lifestyle. Front. Microbiol. 6, 713 (2015).

    Article  Google Scholar 

  40. Burstein, D. et al. Major bacterial lineages are essentially devoid of CRISPR-Cas viral defence systems. Nat. Commun. 7, 10613 (2016).

    Article  CAS  Google Scholar 

  41. Gong, J., Qing, Y., Guo, X. & Warren, A. “Candidatus Sonnebornia yantaiensis”, a member of candidate division OD1, as intracellular bacteria of the ciliated protist Paramecium bursaria (Ciliophora, Oligohymenophorea). Syst. Appl. Microbiol. 37, 35–41 (2014).

    Article  CAS  Google Scholar 

  42. Soro, V. et al. Axenic culture of a candidate division TM7 bacterium from the human oral cavity and biofilm interactions with other oral bacteria. Appl. Environ. Microbiol. 80, 6480–6489 (2014).

    Article  Google Scholar 

  43. He, X. et al. Cultivation of a human-associated TM7 phylotype reveals a reduced genome and epibiotic parasitic lifestyle. Proc. Natl. Acad. Sci. USA 112, 244–249 (2015).

    Article  CAS  Google Scholar 

  44. Luo, F., Devine, C.E. & Edwards, E.A. Cultivating microbial dark matter in benzene-degrading methanogenic consortia. Environ. Microbiol. 18, 2923–2936 (2016).

    Article  CAS  Google Scholar 

  45. Vieira-Silva, S. & Rocha, E.P.C. The systemic imprint of growth and its uses in ecological (meta)genomics. PLoS Genet. 6, e1000808 (2010).

    Article  Google Scholar 

  46. Carini, P. et al. Relic DNA is abundant in soil and obscures estimates of soil microbial diversity. Preprint at bioRxiv http://dx.doi.org/10.1101/043372 (2016).

  47. Langmead, B. & Salzberg, S.L. Fast gapped-read alignment with Bowtie 2. Nat. Methods 9, 357–359 (2012).

    Article  CAS  Google Scholar 

  48. Newville, M., Stensitzki, T., Allen, D.B. & Ingargiola, A. LMFIT: non-linear least-square minimization and curve-fitting for Python (Zenodo, 2014).

  49. Grigoriev, A. Analyzing genomes with cumulative skew diagrams. Nucleic Acids Res. 26, 2286–2290 (1998).

    Article  CAS  Google Scholar 

  50. Ross, M.G. et al. Characterizing and measuring bias in sequence data. Genome Biol. 14, R51 (2013).

    Article  Google Scholar 

  51. Richter, M. & Rosselló-Móra, R. Shifting the genomic gold standard for the prokaryotic species definition. Proc. Natl. Acad. Sci. USA 106, 19126–19131 (2009).

    Article  CAS  Google Scholar 

  52. Altschul, S.F., Gish, W., Miller, W., Myers, E.W. & Lipman, D.J. Basic local alignment search tool. J. Mol. Biol. 215, 403–410 (1990).

    Article  CAS  Google Scholar 

  53. Rissman, A.I. et al. Reordering contigs of draft genomes using the Mauve aligner. Bioinformatics 25, 2071–2073 (2009).

    Article  CAS  Google Scholar 

  54. Ondov, B.D. et al. Mash: fast genome and metagenome distance estimation using MinHash. Genome Biol. 17, 132 (2016).

    Article  Google Scholar 

  55. Raes, J., Korbel, J.O., Lercher, M.J., von Mering, C. & Bork, P. Prediction of effective genome size in metagenomic samples. Genome Biol. 8, R10 (2007).

    Article  Google Scholar 

Download references

Acknowledgements

Funding was provided by NIH grant R01AI092531 Sloan Foundation grant APSF-2012-10-05, and by the US Department of Energy (DOE), Office of Science, Office of Biological and Environmental Research under award number DE-AC02-05CH11231 (Sustainable Systems Scientific Focus Area and DOE-JGI) and award number DE-SC0004918 (Systems Biology Knowledge Base Focus Area). We thank T. Raveh-Sadka, B. Brooks, and D. Burstein for helpful discussions, and M. Albertsen for comments regarding GC sequencing bias.

Author information

Authors and Affiliations

Authors

Contributions

C.T.B. and J.F.B. developed the iRep and bPTR methods. M.R.O. ordered and oriented draft genome sequences for bPTR calculations and conducted kPTR analyses. C.T.B. conducted the iRep, bPTR, and kPTR comparisons, and determined the accuracy of the iRep method. J.F.B. binned the adult human metagenome and curated the Deltaproteobacterium genome, with input from C.T.B. C.T.B. implemented the iRep method. B.C.T. provided bioinformatics support. C.T.B. and J.F.B. drafted the manuscript. All authors contributed to iRep development, reviewed results, and approved the manuscript.

Corresponding author

Correspondence to Jillian F Banfield.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Integrated supplementary information

Supplementary Figure 1 Schematic showing steps involved in a genome-resolved metagenomics study that includes iRep analysis.

Microbiome sample collection and DNA extraction methods should be determined on a per-project basis, and metagenome sequencing can be conducted on the Illumina, PacBio, or another sequencing platform. Sequencing reads are trimmed based on quality scores (e.g. using SickleSickle18) and filtered for contamination (e.g. removal of human genome sequences). High-quality reads are then assembled (e.g. using IDBA_UDUD19), and the resulting scaffolds are binned either manually (e.g. based on GC content, taxonomic affiliation, coverage), and/or using a clustering algorithm such as ESOMESOM20,29,30) or using an automated binning program (e.g. MaxBinMaxBin21, CONCOCTCONCOCT22, or ABAWACAABAWACA15). Genome bins can then be assessed for completion and contamination based on inventory of expected single copy genes (SCGs), either based on identification of these genes from genome annotations (seesee15,29,55), or using software such as CheckMCheckM23. High-quality genomes are then compared with one another and grouped into clusters based on average nucleotide identity (ANI; e.g., based on sharing 98% ANI determined using MashMash54). A representative of each cluster should be included in a genome database that will be used for iRep analysis, along with genomes from other projects that may be appropriate for the analysis. Reads from each metagenome are then mapped to the genome database (e.g. using Bowtie2Bowtie247), and iRep is calculated from the read mapping data (see Online Methods).

Supplementary Figure 2 Evaluation of iRep method parameters.

(a) Gamma distribution used to simulate genome fragmentation for genome completeness analyses. The frequency of genome fragment sizes from all genomes analyzed in this study are compared with genome fragment sizes simulated using a gamma distribution with parameters: alpha = 0.1, beta = 21,000, min. = 5,000, max. = 200,000. These parameters were first estimated by fitting to the genome data, and then manually adjusted. Similarity between the two distributions shows that this gamma distribution can be used to approximate the level of genome fragmentation expected for draft-quality genome sequences. (b) iRep was calculated from random genome fragmentation simulations in order to survey a range of fragmentation levels (Supplementary Table 1). The analysis was conducted for an L. gasseri sample from the Korem et al.8 study in which iRep was determined to be 2.01 using the complete genome with 25x sequencing coverage. This known iRep value was then compared with iRep values determined from each genome fragmentation simulation after subsampling to 75% of the genome and using only 5x sequencing coverage. This enabled analysis of the influence of fragmentation on iRep calculations at the completeness and coverage limits of the method. Results show that 91.8% of iRep values are within the expected range of 0.15 when genomes have fewer than 175 fragments/Mbp of genome sequence. (c) Four L. gasseri samples from the Korem et al.8 study that represent iRep values between 1.50 and 2.01 were selected in order to test different coverage sliding window calculation methods (see Online Methods for description of each methods) and window sizes. For each sample, 100 random genome fragmentations and subsets were conducted in order to assess each method based on various levels of genome completion. The results show that the “iRep” and “median iRep” methods using 5 Kbp windows exhibited the least amount of variation. (d) Because the iRep method involves randomly combining coverage data from different genome fragments prior to calculating coverage sliding windows, some sliding windows will include coverage values from different locations on the complete genome sequence. In order to evaluate the variation introduced by the (random) order in which scaffolds are combined, iRep calculations were conducted for ten random orderings of 100 random genome fragmentations conducted using the sample set described in (c). Results show a very minimal amount of variation in iRep values as described by the difference between the lowest and highest values determined from each of the ten orderings (“iRep range”). Because of this, we chose not to implement the “median iRep” strategy. (e) Using the sample set described in (c), the iRep method was implemented using 5 Kbp windows using different window slide values in order to test whether or not the slide value would change the results. Because both 10 and 100 bp window slides produced similar results, we implemented the iRep method using a 100 bp window slide. (f) iRep is not as strongly correlated with bPTR without the GC sequencing bias correction for five genome sequences assembled from premature infant metagenomes (Supplementary Table 4; compare with GC corrected data in Fig. 2e).

Supplementary Figure 3 Coverage, GC skew patterns, and bPTR measurements for reconstructed genomes oriented and ordered based on complete reference genome sequences.

(a-e) Read mapping was conducted using sequences from the sample used for genome recovery. bPTR was calculated after determining the origin and terminus of replication based on cumulative GC skew. Coverage was calculated for 10 Kbp windows calculated every 100 bp (extremely low and high coverage windows were filtered out; see Online Methods). bPTR was calculated as the ratio between the coverage at the origin and terminus after applying a median filter. Cumulative GC skew and coverage patterns confirm the ordering of genome fragments.

Supplementary Figure 4 Reference genomes are not representative of organisms surveyed in the premature infant microbiome study.

Reads were mapped to both reconstructed genomes and closely related reference genomes (Supplementary Table 4), and the percent of each genome covered by sequencing reads is reported. Average nucleotide identity (ANI) is reported between each reconstructed genome and the paired reference genome. The large fractions of reference genomes not represented by metagenome sequencing show that extensive genomic variation is present between surveyed and reference genomes, despite high ANI values in some cases.

Supplementary Figure 5 Replication rates determined by iRep and kPTR are not in strong agreement for the premature infant study.

iRep values were determined based on reconstructed genomes, and kPTR values based on complete reference genomes (r = Pearson’s r value; Supplementary Tables 5 and 8).

Supplementary Figure 6 Coverage, cumulative GC skew, and bPTR measurements for complete reference genomes with similarity to genomes from the adult human microbiome sample.

(a-e) Reads from the adult human microbiome were mapped to complete reference genome sequences. Coverage was calculated for 10 Kbp windows every 100 bp (extremely low and high coverage windows were filtered out; see Online Methods). The origin and terminus of replication were determined based on coverage. bPTR was calculated as the ratio between the coverage at the origin and terminus after applying a median filter. Cumulative GC skew and coverage patterns suggest the presence of genomic variation or assembly errors for some genomes (b-c, e).

Supplementary Figure 7 Absolute abundance (bars, left axis) and iRep (scatter plot, right axis) for bacterial species associated with premature infants.

The five days following antibiotic administration are indicated using a color gradient (DOL = day of life). Half of the infants in the study developed necrotizing enterocolitis (NEC; dotted red lines) during the study period.

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–7 (PDF 9721 kb)

Supplementary Table 1

Analysis of the impact of genome completeness on iRep replication rate measurements. (XLSX 6986 kb)

Supplementary Table 2

Comparison of iRep, bPTR, and kPTR measurements. (XLSX 47 kb)

Supplementary Table 3

iRep, bPTR, and kPTR measurements for minimum genome sequencing coverage analyses. (XLSX 69 kb)

Supplementary Table 4

Comparison of iRep and bPTR measurements for draft-qualitygenomes ordered and oriented based on complete genome sequences. (XLSX 87 kb)

Supplementary Table 5

iRep measurements for organisms associated with prematureinfant microbiomes. (XLSX 163 kb)

Supplementary Table 6

Single copy gene inventory for genomes reconstructed from anadult human gut metagenome. (XLSX 38 kb)

Supplementary Table 7

iRep measurements for organisms associated with an adulthuman microbiome. (XLSX 48 kb)

Supplementary Table 8

kPTR values determined from the premature infantmetagenomes. (XLSX 46 kb)

Supplementary Table 9

kPTR values determined from the adult human metagenome. (XLSX 33 kb)

Supplementary Table 10

iRep measurements for Candidate Phyla Radiation (CPR)organisms. (XLSX 199 kb)

Supplementary Code (ZIP 89004 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Brown, C., Olm, M., Thomas, B. et al. Measurement of bacterial replication rates in microbial communities. Nat Biotechnol 34, 1256–1263 (2016). https://doi.org/10.1038/nbt.3704

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nbt.3704

This article is cited by

Search

Quick links

Nature Briefing Microbiology

Sign up for the Nature Briefing: Microbiology newsletter — what matters in microbiology research, free to your inbox weekly.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing: Microbiology