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.

  • Letter
  • Published:

Multiplex single-molecule interaction profiling of DNA-barcoded proteins

Abstract

In contrast with advances in massively parallel DNA sequencing1, high-throughput protein analyses2,3,4 are often limited by ensemble measurements, individual analyte purification and hence compromised quality and cost-effectiveness. Single-molecule protein detection using optical methods5 is limited by the number of spectrally non-overlapping chromophores. Here we introduce a single-molecular-interaction sequencing (SMI-seq) technology for parallel protein interaction profiling leveraging single-molecule advantages. DNA barcodes are attached to proteins collectively via ribosome display6 or individually via enzymatic conjugation. Barcoded proteins are assayed en masse in aqueous solution and subsequently immobilized in a polyacrylamide thin film to construct a random single-molecule array, where barcoding DNAs are amplified into in situ polymerase colonies (polonies)7 and analysed by DNA sequencing. This method allows precise quantification of various proteins with a theoretical maximum array density of over one million polonies per square millimetre. Furthermore, protein interactions can be measured on the basis of the statistics of colocalized polonies arising from barcoding DNAs of interacting proteins. Two demanding applications, G-protein coupled receptor and antibody-binding profiling, are demonstrated. SMI-seq enables ‘library versus library’ screening in a one-pot assay, simultaneously interrogating molecular binding affinity and specificity.

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: Schematics of protein barcoding methods.
Figure 2: Amplification and quantification of barcoding DNAs.
Figure 3: Analyses of protein interactions via polony colocalization.
Figure 4: Parallel antibody binding profiling.

Similar content being viewed by others

References

  1. Shendure, J. & Ji, H. Next-generation DNA sequencing. Nature Biotechnol. 26, 1135–1145 (2008)

    CAS  Google Scholar 

  2. Dreze, M. et al. High-quality binary interactome mapping. Methods Enzymol. 470, 281–315 (2010)

    CAS  PubMed  Google Scholar 

  3. MacBeath, G. & Schreiber, S. L. Printing proteins as microarrays for high-throughput function determination. Science 289, 1760–1763 (2000)

    ADS  CAS  PubMed  Google Scholar 

  4. Gavin, A. C. et al. Functional organization of the yeast proteome by systematic analysis of protein complexes. Nature 415, 141–147 (2002)

    ADS  CAS  PubMed  Google Scholar 

  5. Weiss, S. Fluorescence spectroscopy of single biomolecules. Science 283, 1676–1683 (1999)

    ADS  CAS  PubMed  Google Scholar 

  6. Hanes, J. & Pluckthun, A. In vitro selection and evolution of functional proteins by using ribosome display. Proc. Natl Acad. Sci. USA 94, 4937–4942 (1997)

    ADS  CAS  PubMed  PubMed Central  Google Scholar 

  7. Mitra, R. D. & Church, G. M. In situ localized amplification and contact replication of many individual DNA molecules. Nucleic Acids Res. 27, e34–e39 (1999)

    CAS  PubMed  PubMed Central  Google Scholar 

  8. Shimizu, Y. et al. Cell-free translation reconstituted with purified components. Nature Biotechnol. 19, 751–755 (2001)

    CAS  Google Scholar 

  9. Los, G. V. et al. HatoTag: A novel protein labeling technology for cell imaging and protein analysis. ACS Chem. Biol. 3, 373–382 (2008)

    CAS  PubMed  Google Scholar 

  10. Bentley, D. R. et al. Accurate whole human genome sequencing using reversible terminator chemistry. Nature 456, 53–59 (2008)

    ADS  CAS  PubMed  PubMed Central  Google Scholar 

  11. Shendure, J. et al. Accurate multiplex polony sequencing of an evolved bacterial genome. Science 309, 1728–1732 (2005)

    ADS  CAS  PubMed  Google Scholar 

  12. Aird, D. et al. Analyzing and minimizing PCR amplification bias in Illumina sequencing libraries. Genome Biol. 12, R18 (2011)

    CAS  PubMed  PubMed Central  Google Scholar 

  13. Fredriksson, S. et al. Protein detection using proximity-dependent DNA ligation assays. Nature Biotechnol. 20, 473–477 (2002)

    CAS  Google Scholar 

  14. Hammond, M., Nong, R. Y., Ericsson, O., Pardali, K. & Landegren, U. Profiling cellular protein complexes by proximity ligation with dual tag microarray readout. PLoS ONE 7, e40405 (2012)

    ADS  CAS  PubMed  PubMed Central  Google Scholar 

  15. Lundberg, M., Eriksson, A., Tran, B., Assarsson, E. & Fredriksson, S. Homogeneous antibody-based proximity extension assays provide sensitive and specific detection of low-abundant proteins in human blood. Nucleic Acids Res. 39, e102 (2011)

    CAS  PubMed  PubMed Central  Google Scholar 

  16. Zhu, J. et al. Protein interaction discovery using parallel analysis of translated ORFs (PLATO). Nature Biotechnol. 31, 331–334 (2013)

    CAS  Google Scholar 

  17. Vetter, I. R. & Wittinghofer, A. The guanine nucleotide-binding switch in three dimensions. Science 294, 1299–1304 (2001)

    ADS  CAS  PubMed  Google Scholar 

  18. Block, C., Janknecht, R., Herrmann, C., Nassar, N. & Wittinghofer, A. Quantitative structure-activity analysis correlating Ras/Raf interaction in vitro to Raf activation in vivo. Nature Struct. Biol. 3, 244–251 (1996)

    CAS  PubMed  Google Scholar 

  19. Kiel, C. et al. Improved binding of Raf to Ras·GDP is correlated with biological activity. J. Biol. Chem. 284, 31893–31902 (2009)

    CAS  PubMed  PubMed Central  Google Scholar 

  20. Overington, J. P., Al-Lazikani, B. & Hopkins, A. L. How many drug targets are there? Nature Rev. Drug Discov. 5, 993–996 (2006)

    CAS  Google Scholar 

  21. Zhang, R. & Xie, X. Tools for GPCR drug discovery. Acta Pharmacol. Sin. 33, 372–384 (2012)

    CAS  PubMed  PubMed Central  Google Scholar 

  22. Denisov, I. G., Grinkova, Y. V., Lazarides, A. A. & Sligar, S. G. Directed self-assembly of monodisperse phospholipid bilayer nanodiscs with controlled size. J. Am. Chem. Soc. 126, 3477–3487 (2004)

    CAS  PubMed  Google Scholar 

  23. Leitz, A. J., Bayburt, T. H., Barnakov, A. N., Springer, B. A. & Sligar, S. G. Functional reconstitution of β2-adrenergic receptors utilizing self-assembling nanodisc technology. Biotechniques 40, 601–612 (2006)

    CAS  PubMed  Google Scholar 

  24. Whorton, M. R. et al. A monomeric G protein-coupled receptor isolated in a high-density lipoprotein particle efficiently activates its G protein. Proc. Natl Acad. Sci. USA 104, 7682–7687 (2007)

    ADS  CAS  PubMed  PubMed Central  Google Scholar 

  25. Luttrell, L. M. & Lefkowitz, R. J. The role of β-arrestins in the termination and transduction of G-protein-coupled receptor signals. J. Cell Sci. 115, 455–465 (2002)

    CAS  PubMed  Google Scholar 

  26. Gurevich, V. V. et al. Arrestin interactions with G-protein-coupled receptors - Direct binding-studies of wild-type and mutant arrestins with rhodopsin, β2-adrenergic, and m2-muscarinic cholinergic receptors. J. Biol. Chem. 270, 720–731 (1995)

    CAS  PubMed  Google Scholar 

  27. Yu, H. et al. Next-generation sequencing to generate interactome datasets. Nature Methods 8, 478–480 (2011)

    CAS  PubMed  PubMed Central  Google Scholar 

  28. Michaud, G. A. et al. Analyzing antibody specificity with whole proteome microarrays. Nature Biotechnol. 21, 1509–1512 (2003)

    CAS  Google Scholar 

  29. Kosuri, S. et al. Scalable gene synthesis by selective amplification of DNA pools from high-fidelity microchips. Nature Biotechnol. 28, 1295–1299 (2010)

    CAS  Google Scholar 

  30. McGregor, L. M., Jain, T. & Liu, D. R. Identification of ligand-target pairs from combined libraries of small molecules and unpurified protein targets in cell lysates. J. Am. Chem. Soc. 136, 3264–3270 (2014)

    CAS  PubMed  PubMed Central  Google Scholar 

  31. Yang, X. et al. A public genome-scale lentiviral expression library of human ORFs. Nature Methods 8, 659–661 (2011)

    CAS  PubMed  PubMed Central  Google Scholar 

  32. Parker, E. M., Kameyama, K., Higashijima, T. & Ross, E. M. Reconstitutively active G protein-coupled receptors purified from baculovirus-infected insect cells. J. Biol. Chem. 266, 519–527 (1991)

    CAS  PubMed  Google Scholar 

  33. Kobilka, B. K. Amino and carboxyl terminal modifications to facilitate the production and purification of a G protein-coupled receptor. Anal. Biochem. 231, 269–271 (1995)

    CAS  PubMed  Google Scholar 

  34. Mitra, N. et al. Calcium-dependent ligand binding and G-protein signaling of family B GPCR parathyroid hormone 1 receptor purified in nanodiscs. ACS Chem. Biol. 8, 617–625 (2013)

    CAS  PubMed  PubMed Central  Google Scholar 

  35. John, J., Frech, M. & Wittinghofer, A. Biochemical properties of Ha-ras encoded p21 mutants and mechanism of the autophosphorylation reaction. J. Biol. Chem. 263, 11792–11799 (1988)

    CAS  PubMed  Google Scholar 

  36. Mitra, R. D. et al. Digital genotyping and haplotyping with polymerase colonies. Proc. Natl Acad. Sci. USA 100, 5926–5931 (2003)

    ADS  CAS  PubMed  PubMed Central  Google Scholar 

  37. Mitra, R. D., Shendure, J., Olejnik, J., Edyta Krzymanska, O. & Church, G. M. Fluorescent in situ sequencing on polymerase colonies. Anal. Biochem. 320, 55–65 (2003)

    CAS  PubMed  Google Scholar 

  38. Aach, J. & Church, G. M. Mathematical models of diffusion-constrained polymerase chain reactions: basis of high-throughput nucleic acid assays and simple self-organizing systems. J. Theor. Biol. 228, 31–46 (2004)

    MathSciNet  CAS  PubMed  Google Scholar 

  39. Philimonenko, A. A., Janacek, J. & Hozak, P. Statistical evaluation of colocalization patterns in immunogold labeling experiments. J. Struct. Biol. 132, 201–210 (2000)

    CAS  PubMed  Google Scholar 

  40. Lewis, J. D. et al. Quantitative interactor screening with next-generation sequencing (QIS-Seq) identifies Arabidopsis thaliana MLO2 as a target of the Pseudomonas syringae type III effector HopZ2. BMC Genomics 13, 8 (2012)

    CAS  PubMed  PubMed Central  Google Scholar 

  41. Georgiou, G. et al. The promise and challenge of high-throughput sequencing of the antibody repertoire. Nature Biotechnol. 32, 158–168 (2014)

    CAS  Google Scholar 

  42. Larman, H. B. et al. Autoantigen discovery with a synthetic human peptidome. Nature Biotechnol. 29, 535–541 (2011)

    CAS  Google Scholar 

  43. Fujimori, S. et al. Next-generation sequencing coupled with a cell-free display technology for high-throughput production of reliable interactome data. Sci. Rep. 2, 691 (2012)

    PubMed  PubMed Central  Google Scholar 

  44. Darmanis, S. et al. ProteinSeq: high-performance proteomic analyses by proximity ligation and next generation sequencing. PLoS ONE 6, e25583 (2011)

    ADS  CAS  PubMed  PubMed Central  Google Scholar 

  45. Young, K. et al. Identification of a calcium channel modulator using a high throughput yeast two-hybrid screen. Nature Biotechnol. 16, 946–950 (1998)

    CAS  Google Scholar 

  46. Nishihara, T. et al. Estrogenic activities of 517 chemicals by yeast two-hybrid assay. J. Health Sci. 46, 282–298 (2000)

    CAS  Google Scholar 

  47. Chidley, C., Haruki, H., Pedersen, M. G., Muller, E. & Johnsson, K. A yeast-based screen reveals that sulfasalazine inhibits tetrahydrobiopterin biosynthesis. Nature Chem. Biol. 7, 375–383 (2011)

    CAS  Google Scholar 

  48. Wrighton, N. C. et al. Small peptides as potent mimetics of the protein hormone erythropoietin. Science 273, 458–463 (1996)

    ADS  CAS  PubMed  Google Scholar 

  49. Lowman, H. B. Bacteriophage display and discovery of peptide leads for drug development. Annu. Rev. Biophys. Biomol. Struct. 26, 401–424 (1997)

    CAS  PubMed  Google Scholar 

  50. Leuchowius, K. J. et al. High content screening for inhibitors of protein interactions and post-translational modifications in primary cells by proximity ligation. Mol. Cell. Proteomics 9, 178–183 (2010)

    CAS  PubMed  Google Scholar 

Download references

Acknowledgements

This work was supported by a grant from the US Department of Energy (DE-FG02-02ER63445) to G.M.C. and a grant from the NIH NHGRI (HG001715) to M.V. and D.E.H. L.G. was supported by a postdoctoral fellowship from the Jane Coffin Childs Fund for Medical Research and a grant from Harvard Origins of Life Initiative. We thank W. Harper, L. Pontano Vaites, S. Elledge and J. Zhu for providing plasmids, F. Vigneault for comments on the manuscript, J. Lai and D. Breslau for assistance on imaging setup, and members of the Church and Vidal groups for constructive discussions.

Author information

Authors and Affiliations

Authors

Contributions

L.G. and G.M.C. conceived the technique; L.G. and C.L. performed experiments and analysed data; J.A. built the mathematical model and assisted the colocalization analyses; D.E.H. and M.V. assisted the production of barcoded proteins; L.G., J.A. and G.M.C. wrote the manuscript with help from the other authors.

Corresponding authors

Correspondence to Liangcai Gu or George M. Church.

Ethics declarations

Competing interests

A provisional patent has been filed by the Harvard University Office of Technology Development.

Additional information

MATLAB scripts for imaging analyses, colocalization statistics and mathematical modelling can be found at http://arep.med.harvard.edu/SMI-Seq/.

Extended data figures and tables

Extended Data Figure 1 Improved stability of PRMC complexes generated in a reconstituted E. coli IVT system.

a, Schematic of PRMC complex stability analysis by measuring the relative ratio of barcoding DNA to HaloTag-labelled protein. b, Comparison of PRMC complex stabilities in the E. coli recombinant-factor-reconstituted (PURE) and an E. coli crude extract (S30) IVT system. Nucleic acid degradation or ribosome dissociation can result in the loss of barcoding DNAs. IVT reactions were performed at 37 °C for 30 min and PRMC complexes were further incubated at room temperature for indicated periods of time before the affinity purification and the stability analysis. Means of three independent experiments ± standard deviations.

Extended Data Figure 2 HaloTag-based protein–DNA conjugation.

a, Schematic of the individual barcoding method adaptable to an automatic platform. Fusion proteins bearing an N- or C-terminal HaloTag and the affinity tags were purified and conjugated to a barcoding DNA bearing three different modifications. b, Agarose gel electrophoresis of the barcoding DNA and selected protein–DNA conjugates.

Extended Data Figure 3 Covalent immobilization of barcoding DNAs is required for in situ polony amplification.

a, b, Representative images of polonies amplified from barcoding DNA templates without (a) or with (b) 5′-acrydite modifications. Oversized polonies or polony clusters shown in a resulted from template-drifting-induced multiple seeding events during the amplification.

Extended Data Figure 4 Polony quantification of various barcoded proteins.

a, Plot showing the average number of polonies detected at a single imaging position against the average number of barcoding DNA templates predicted by real-time PCR quantification. Data represent mean values of 100 measurements; error bars, 95% CL. b, Log–log plot of total numbers of polonies detected against dilution factors. Data represent mean values of two technical replicates.

Extended Data Figure 5 Crosslinking efficiency of DsRed is improved by a lysine-rich TolA domain.

a, SDS–PAGE analysis of purified DsRed (Clontech Laboratories) and HaloTag–DsRed–TolA proteins before (lanes 1 and 3) and after (lanes 2 and 4) the crosslinking. 10 µM purified proteins were crosslinked by 1 mM BS(PEG)5 in 20 mM HEPES buffer, pH 8.0, 150 mM KOAc at 4 °C for 1 h. Proteins were stained with Coomassie blue. Only a minor band of the crosslinked dimer was observed for DsRed (lane 2); in contrast, HaloTag–DsRed–TolA was all crosslinked as a tetramer or a trimer (lane 4). Co-purified E. coli proteins (some protein bands below the major band in the lane 3), probably bound to TolA during the purification, and degradation products (due to the hydrolysis of an acylimine bond in the DsRed chromophore) were efficiently crosslinked to HaloTag–DsRed–TolA. b, Comparison of HaloTag-labelled DsRed and mCherry, a monomeric fluorescence protein, crosslinked at different conditions. Proteins labelled with Halo-TMR were analysed by fluorescent gel imaging. Only a minor fraction of HaloTag–mCherry–TolA, a control to show non-specific crosslinking, was crosslinked at increased protein concentrations. Intramolecularly crosslinked proteins show multiple bands or smears corresponding to different quaternary structures of the multidomain proteins stabilized by crosslinking. Bands of the crosslinked trimers show an increased intensity at higher BS(PEG)5 concentrations probably because the primary amine groups on surface are more quickly modified by BS(PEG)5, thus preventing the further crosslinking to form the tetramer.

Extended Data Table 1 Polony quantification of titrated binder proteins and antigens
Extended Data Table 2 ScFvs and human proteins used in the one-pot binding profiling
Extended Data Table 3 Comparison of protein interaction profiling technologies based on nucleic acid barcoding and high-throughput sequencing

Supplementary information

Supplementary Information

This file contains Supplementary Methods, Supplementary Notes, a Supplementary Discussion, Supplementary Figure 1 and Supplementary References. (PDF 678 kb)

Supplementary Table 1

The file contains the Ras and Raf-RBD binding assay. (XLSX 10 kb)

Supplementary Table 2

This file contains the GPCR screening assay. (XLSX 11 kb)

Supplementary Table 3

This file contains the ScFv binding profiling assay. (XLSX 142 kb)

Supplementary Table 4

This file contains the DNA constructs used in this study. (XLSX 31 kb)

Supplementary Table 5

The file contains the Sequences of expression vectors and synthetic oligos used in this study. (XLSX 25 kb)

PowerPoint slides

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Gu, L., Li, C., Aach, J. et al. Multiplex single-molecule interaction profiling of DNA-barcoded proteins. Nature 515, 554–557 (2014). https://doi.org/10.1038/nature13761

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nature13761

This article is cited by

Comments

By submitting a comment you agree to abide by our Terms and Community Guidelines. If you find something abusive or that does not comply with our terms or guidelines please flag it as inappropriate.

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