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:

Single-cell genome sequencing at ultra-high-throughput with microfluidic droplet barcoding

This article has been updated

Abstract

The application of single-cell genome sequencing to large cell populations has been hindered by technical challenges in isolating single cells during genome preparation. Here we present single-cell genomic sequencing (SiC-seq), which uses droplet microfluidics to isolate, fragment, and barcode the genomes of single cells, followed by Illumina sequencing of pooled DNA. We demonstrate ultra-high-throughput sequencing of >50,000 cells per run in a synthetic community of Gram-negative and Gram-positive bacteria and fungi. The sequenced genomes can be sorted in silico based on characteristic sequences. We use this approach to analyze the distributions of antibiotic-resistance genes, virulence factors, and phage sequences in microbial communities from an environmental sample. The ability to routinely sequence large populations of single cells will enable the de-convolution of genetic heterogeneity in diverse cell populations.

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: Schematic of SiC-seq workflow.
Figure 2: Microfluidic and biochemical workflow to generate a SiC-seq library.
Figure 3: SiC-seq performance on an artificial microbial community consisting of ten different cell species.
Figure 4: Application of SiC-seq to a marine community recovered from the San Francisco coastline.

Similar content being viewed by others

Accession codes

Primary accessions

Sequence Read Archive

Referenced accessions

NCBI Reference Sequence

Change history

  • 21 June 2017

    In the version of this article initially published, in Figure 3c, bars on the x axis were labeled S. epidermidis rather than S. enterica. The error has been corrected for the print, PDF and HTML versions of this article.

References

  1. Del Monte, U. Does the cell number 10(9) still really fit one gram of tumor tissue? Cell Cycle 8, 505–506 (2009).

    Article  CAS  Google Scholar 

  2. Maranger, R. & Bird, D. Viral abundance in aquatic systems: a comparison between marine and fresh waters. Mar. Ecol. Prog. Ser. 121, 217–226 (1995).

    Article  Google Scholar 

  3. Zhang, H. & Liu, K.-K. Optical tweezers for single cells. J. R. Soc. Interface 5, 671–690 (2008).

    Article  CAS  Google Scholar 

  4. Rinke, C. et al. Obtaining genomes from uncultivated environmental microorganisms using FACS-based single-cell genomics. Nat. Protoc. 9, 1038–1048 (2014).

    Article  CAS  Google Scholar 

  5. Gawad, C., Koh, W. & Quake, S.R. Dissecting the clonal origins of childhood acute lymphoblastic leukemia by single-cell genomics. Proc. Natl. Acad. Sci. USA 111, 17947–17952 (2014).

    Article  CAS  Google Scholar 

  6. Leung, K. et al. Robust high-performance nanoliter-volume single-cell multiple displacement amplification on planar substrates. Proc. Natl. Acad. Sci. USA 113, 8484–8489 (2016).

    Article  CAS  Google Scholar 

  7. Tamminen, M.V. & Virta, M.P.J. Single gene-based distinction of individual microbial genomes from a mixed population of microbial cells. Front. Microbiol. 6, 195 (2015).

    Article  Google Scholar 

  8. 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 

  9. Xu, L., Brito, I.L., Alm, E.J. & Blainey, P.C. Virtual microfluidics for digital quantification and single-cell sequencing. Nat. Methods 13, 759–762 (2016).

    Article  CAS  Google Scholar 

  10. Lan, F., Haliburton, J.R., Yuan, A. & Abate, A.R. Droplet barcoding for massively parallel single-molecule deep sequencing. Nat. Commun. 7, 11784 (2016).

    Article  CAS  Google Scholar 

  11. Rotem, A. et al. Single-cell ChIP-seq reveals cell subpopulations defined by chromatin state. Nat. Biotechnol. 33, 1165–1172 (2015).

    Article  CAS  Google Scholar 

  12. Klein, A.M. et al. Droplet barcoding for single-cell transcriptomics applied to embryonic stem cells. Cell 161, 1187–1201 (2015).

    Article  CAS  Google Scholar 

  13. Macosko, E.Z. et al. Highly parallel genome-wide expression profiling of individual cells using nanoliter droplets. Cell 161, 1202–1214 (2015).

    Article  CAS  Google Scholar 

  14. Rotem, A. et al. High-throughput single-cell labeling (Hi-SCL) for RNA-seq using drop-based microfluidics. PLoS One 10, e0116328 (2015).

    Article  Google Scholar 

  15. Novak, R. et al. Single-cell multiplex gene detection and sequencing with microfluidically generated agarose emulsions. Angew. Chem. Int. Edn Engl. 50, 390–395 (2011).

    Article  CAS  Google Scholar 

  16. Garstecki, P. et al. Formation of monodisperse bubbles in a microfluidic flow-focusing device. Appl. Phys. Lett. 85, 2649–2651 (2004).

    Article  CAS  Google Scholar 

  17. Amini, S. et al. Haplotype-resolved whole-genome sequencing by contiguity-preserving transposition and combinatorial indexing. Nat. Genet. 46, 1343–1349 (2014).

    Article  CAS  Google Scholar 

  18. de Bourcy, C.F.A. et al. A quantitative comparison of single-cell whole genome amplification methods. PLoS One 9, e105585 (2014).

    Article  Google Scholar 

  19. Gole, J. et al. Massively parallel polymerase cloning and genome sequencing of single cells using nanoliter microwells. Nat. Biotechnol. 31, 1126–1132 (2013).

    Article  CAS  Google Scholar 

  20. Nathan, C. & Cars, O. Antibiotic resistance--problems, progress, and prospects. N. Engl. J. Med. 371, 1761–1763 (2014).

    Article  Google Scholar 

  21. Yildiz, F.H. Processes controlling the transmission of bacterial pathogens in the environment. Res. Microbiol. 158, 195–202 (2007).

    Article  Google Scholar 

  22. Jiang, S.C. & Paul, J.H. Gene transfer by transduction in the marine environment. Appl. Environ. Microbiol. 64, 2780–2787 (1998).

    CAS  PubMed  PubMed Central  Google Scholar 

  23. Ochman, H. & Moran, N.A. Genes lost and genes found: evolution of bacterial pathogenesis and symbiosis. Science 292, 1096–1099 (2001).

    Article  CAS  Google Scholar 

  24. Human Microbiome Project Consortium. Structure, function and diversity of the healthy human microbiome. Nature 486, 207–214 (2012).

  25. Afshinnekoo, E. et al. Geospatial resolution of human and bacterial diversity with city-scale metagenomics. Cell Syst. 1, 72–87 (2015).

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

  27. Ni, X. et al. Reproducible copy number variation patterns among single circulating tumor cells of lung cancer patients. Proc. Natl. Acad. Sci. USA 110, 21083–21088 (2013).

    Article  CAS  Google Scholar 

  28. Liu, B. & Pop, M. ARDB—Antibiotic Resistance Genes Database. Nucleic Acids Res. 37 (Suppl. 1), D443–D447 (2009).

    Article  CAS  Google Scholar 

  29. Chen, L., Xiong, Z., Sun, L., Yang, J. & Jin, Q. VFDB 2012 update: toward the genetic diversity and molecular evolution of bacterial virulence factors. Nucleic Acids Res. 40, D641–D645 (2012).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We are grateful for K. Stedman, R. Malmstrom, R. Andino, K. Pollard, and M. Fischbach for very helpful discussion of and advice on the manuscript. We thank C. O'Loughlin at UCSF for providing microbial strains. This work was supported by the National Science Foundation through a CAREER Award (grant number DBI-1253293); the National Institutes of Health (NIH) (grant numbers HG007233-01, R01-EB019453-01, 1R21HG007233, DP2-AR068129-01, R01-HG008978); and the Defense Advanced Research Projects Agency Living Foundries Program (contract numbers HR0011-12-C-0065, N66001-12-C-4211, HR0011-12-C-0066). Funding for open access charge: (NIH grant number DP2-AR068129-01). F.L. is supported by a PGS-D grant from the National Science and Engineering Research Council of Canada (NSERC).

Author information

Authors and Affiliations

Authors

Contributions

F.L. and A.R.A. conceived of the SiC-seq method. F.L., B.D., and N.A. designed and performed the experiments, and analyzed data. F.L. and A.R.A. wrote the manuscript.

Corresponding author

Correspondence to Adam R Abate.

Ethics declarations

Competing interests

Patents pertaining to this workflow may be licensed to Mission Bio, of which A.R.A. is a shareholder.

Integrated supplementary information

Supplementary Figure 1 Drawings of microfluidic devices.

Schematics of microfluidic devices used to: a) generate barcode droplets and encapsulate cells in agarose microgels; b) re-encapsulate gels in tagmentation reagents; c) merge gel droplets with barcode droplets and PCR droplets.

Supplementary Figure 2 Characterizing diffusion of genomic DNA fragments in agarose microgels

a) Bacterial genomes were stained with SYBR Green I to visually monitor diffusion of genomes in microgels before and after tagmentation. b) Bioanalyzer traces of samples taken from microgel contents and the supernatant. The shift in fragment size is relatively minor as a result of the relatively low stoichiometric ratio of transposase to genome used. c) Bioanalyzer traces from microgel-encapsulated genomes reacted with a higher stoichiometric ratio of transposase to genome. After 1 hr, a peak centered at ~150 bp representing tagmented DNA is visible for the sample containing tagmention enzyme.

Supplementary Figure 3 Lorenz curves of barcode group coverage, by species.

The average depth and distribution of genome coverage of each barcode group plotted as a Lorenz curve for each species in the 10-cell control experiment.

Supplementary Figure 4 Genome size-normalized barcode group purity scores.

Genome size-normalized purity scores of barcode groups in the 10-cell control experiment. Genome size-normalized purity scores are calculated using the same method using the fraction of the genome sequenced for each respective species rather than the raw number of reads.

Supplementary Figure 5 Barcode group purities, by species.

Purity scores of barcode groups separately plotted for each species in the 10-cell control experiment.

Supplementary Figure 6 Barcode group purity scores for second-most abundant species.

Purity scores of the next-most abundant species in a) barcode groups of purity <80% and b) barcode groups of purity >80%. In barcode groups with <80% purity, the purity scores of the next-most abundant species tend to be high from ~20% to 50%, reflecting that those two species represent the majority of the reads in the barcode group, suggesting that these barcode groups represent double encapsulations. Barcode groups with 100% purity are not represented in the plots. Blue line represents cumulative barcode counts normalized to 1.

Supplementary Figure 7 SiC-seq performance on an artificially constructed microbial community consisting of Staphylococcus, Bacillus, and Saccharomyces.

Relative abundance estimates of each species are calculated using barcode counting (Barcode), marker gene counting without barcodes (Metaphlan), and manual counting under the microscope after cell encapsulation (Microscope count) and while in culture (Theoretical).

Supplementary Figure 8 Aggregate genomic coverage of all the barcode groups for species in the synthetic microbial community.

Species at low abundance show frequent dropouts characterized by dips in the graph, but instances of systematic bias characterized by sharp peaks are rarely observed.

Supplementary Figure 9 Read distribution across the Staphylococcus genome for individual barcode groups.

Genomic mapping positions of reads for randomly chosen individual barcode Staphylococcus groups with >2000 reads.

Supplementary Figure 10 Schematic depiction of the framework of the SiC-Reads database.

SiC-seq reads which contain properties such as sequence, read ID, and taxonomy are stored inside barcode groups which contain properties such as purity and taxonomy.

Supplementary Figure 11 Analysis of the marine microbial community used to demonstrate in silico cytometry.

a) Taxonomic abundance of the SiC-Reads database by barcode groups; b) Distribution of purity of barcode groups in the database at the genus level.

Supplementary Figure 12 Reference data obtained by simulating reads from genomic sequences of isolated strains for comparison against data in the marine microbial community.

a) Antibiotic resistance network for whole genome sequenced strains in public databases; b) Virulence factor ratios calculated for publically available strains.

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–12 (PDF 5470 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Lan, F., Demaree, B., Ahmed, N. et al. Single-cell genome sequencing at ultra-high-throughput with microfluidic droplet barcoding. Nat Biotechnol 35, 640–646 (2017). https://doi.org/10.1038/nbt.3880

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nbt.3880

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