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:

Quantification of extracellular proteins, protein complexes and mRNAs in single cells by proximity sequencing

Abstract

We present proximity sequencing (Prox-seq) for simultaneous measurement of proteins, protein complexes and mRNAs in thousands of single cells. Prox-seq combines proximity ligation assay with single-cell sequencing to measure proteins and their complexes from all pairwise combinations of targeted proteins, providing quadratically scaled multiplexing. We validate Prox-seq and analyze a mixture of T cells and B cells to show that it accurately identifies these cell types and detects well-known protein complexes. Next, by studying human peripheral blood mononuclear cells, we discover that naïve CD8+ T cells display the protein complex CD8–CD9. Finally, we study protein interactions during Toll-like receptor (TLR) signaling in human macrophages. We observe the formation of signal-specific protein complexes, find CD36 co-receptor activity and additive signal integration under lipopolysaccharide (TLR4) and Pam2CSK4 (TLR2) stimulation, and show that quantification of protein complexes identifies signaling inputs received by macrophages. Prox-seq provides access to an untapped measurement modality for single-cell phenotyping and can discover uncharacterized protein interactions in different cell types.

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: Overview of Prox-seq for joint proteomic and transcriptomic analysis of single cells.
Fig. 2: Quantification of protein complexes and the proximity ligation background.
Fig. 3: Prox-seq reveals a new CD9–CD8 interaction in peripheral blood mononuclear cells.
Fig. 4: Simultaneous protein and mRNA measurements by Prox-seq on 8,700 single peripheral blood mononuclear cells.
Fig. 5: Prox-seq enables the study of receptor interactions under combined Toll-like receptor stimulation in macrophages.
Fig. 6: Prox-seq reveals single-macrophage variability in TLR signaling and enables identification of immune inputs from protein measurements.

Similar content being viewed by others

Data availability

The raw and count data are deposited in NCBI’s Gene Expression Omnibus under accession numbers GSE149574 and GSE196130. Source data are provided with this paper.

Code availability

The custom program for PLA product alignment and the codes used for alignment and data analysis are available at https://github.com/tay-lab/Prox-seq/.

References

  1. Grün, D. et al. Single-cell messenger RNA sequencing reveals rare intestinal cell types. Nature 525, 251–255 (2015).

    Google Scholar 

  2. Rizvi, A. H. et al. Single-cell topological RNA-seq analysis reveals insights into cellular differentiation and development. Nat. Biotechnol. 35, 551–560 (2017).

    CAS  Google Scholar 

  3. Tay, S. et al. Single-cell NF-κB dynamics reveal digital activation and analogue information processing. Nature 466, 267–271 (2010).

    CAS  Google Scholar 

  4. Söderberg, O. et al. Direct observation of individual endogenous protein complexes in situ by proximity ligation. Nat. Methods 3, 995–1000 (2006).

    Google Scholar 

  5. Stoeckius, M. et al. Simultaneous epitope and transcriptome measurement in single cells. Nat. Methods 14, 865–868 (2017).

    CAS  Google Scholar 

  6. Peterson, V. M. et al. Multiplexed quantification of proteins and transcripts in single cells. Nat. Biotechnol. 35, 936–939 (2017).

    CAS  Google Scholar 

  7. Chi, Q., Wang, G. & Jiang, J. The persistence length and length per base of single-stranded DNA obtained from fluorescence correlation spectroscopy measurements using mean field theory. Phys. A: Stat. Mech. Appl. 392, 1072–1079 (2013).

    CAS  Google Scholar 

  8. Birnbaum, M. E. et al. Molecular architecture of the αβ T cell receptor–CD3 complex. Proc. Natl Acad. Sci. USA 111, 17576–17581 (2014).

    CAS  Google Scholar 

  9. Picelli, S. et al. Full-length RNA-seq from single cells using Smart-seq2. Nat. Protoc. 9, 171–181 (2014).

    CAS  Google Scholar 

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

    CAS  Google Scholar 

  11. Albayrak, C. et al. Digital quantification of proteins and mRNA in single mammalian cells. Mol. Cell 61, 914–924 (2016).

    CAS  Google Scholar 

  12. Lin, J. et al. Ultra-sensitive digital quantification of proteins and mRNA in single cells. Nat. Commun. 10, 3544 (2019).

    Google Scholar 

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

    CAS  Google Scholar 

  14. Genshaft, A. S. et al. Multiplexed, targeted profiling of single-cell proteomes and transcriptomes in a single reaction. Genome Biol. 17, 188 (2016).

    Google Scholar 

  15. Darmanis, S. et al. Simultaneous multiplexed measurement of RNA and proteins in single cells. Cell Rep. 14, 380–389 (2016).

    CAS  Google Scholar 

  16. Leuchowius, K.-J. et al. Parallel visualization of multiple protein complexes in individual cells in tumor tissue. Mol. Cell. Proteomics 12, 1563–1571 (2013).

    CAS  Google Scholar 

  17. Ziegenhain, C. et al. Comparative analysis of single-cell RNA-sequencing methods. Mol. Cell 65, 631–643 (2017).

    CAS  Google Scholar 

  18. Alsemarz, A., Lasko, P. & Fagotto, F. Limited significance of the in situ proximity ligation assay. Preprint at bioRxiv https://doi.org/10.1101/411355 (2018).

  19. Van Der Merwe, P. A. & Dushek, O. Mechanisms for T cell receptor triggering. Nat. Rev. Immunol. 11, 47–55 (2011).

    Google Scholar 

  20. Esensten, J. H., Helou, Y. A., Chopra, G., Weiss, A. & Bluestone, J. A. CD28 costimulation: from mechanism to therapy. Immunity 44, 973–988 (2016).

    CAS  Google Scholar 

  21. Zak, K. M. et al. Structural biology of the immune checkpoint receptor PD-1 and its ligands PD-L1/PD-L2. Structure 25, 1163–1174 (2017).

    CAS  Google Scholar 

  22. Cherry, R. J. et al. Detection of dimers of dimers of human leukocyte antigen (HLA)-DR on the surface of living cells by single-particle fluorescence imaging. J. Cell Biol. 140, 71–79 (1998).

    CAS  Google Scholar 

  23. Cochran, J. R., Cameron, T. O. & Stern, L. J. The relationship of MHC-peptide binding and T cell activation probed using chemically defined MHC class II oligomers. Immunity 12, 241–250 (2000).

    CAS  Google Scholar 

  24. Bhatia, S., Edidin, M., Almo, S. C. & Nathenson, S. G. Different cell surface oligomeric states of B7-1 and B7-2: Implications for signaling. Proc. Natl Acad. Sci. USA 102, 15569–15574 (2005).

    CAS  Google Scholar 

  25. Reilly, P. L. et al. The native structure of intercellular adhesion molecule-1 (ICAM-1) is a dimer. Correlation with binding to LFA-1. J. Immunol. 155, 529–532 (1995).

    CAS  Google Scholar 

  26. Kovalenko, O. V., Yang, X., Kolesnikova, T. V. & Hemler, M. E. Evidence for specific tetraspanin homodimers: inhibition of palmitoylation makes cysteine residues available for cross-linking. Biochem. J. 377, 407–417 (2004).

    CAS  Google Scholar 

  27. Thome, M., Germain, V., Disanto, J. P. & Acuto, O. The p56lck SH2 domain mediates recruitment of CD8/p56lck to the activated T cell receptor/CD3/ζ complex. Eur. J. Immunol. 26, 2093–2100 (1996).

    CAS  Google Scholar 

  28. Collins, T. L. et al. p56lck association with CD4 is required for the interaction between CD4 and the TCR/CD3 complex and for optimal antigen stimulation. J. Immunol. 148, 2159–2162 (1992).

    CAS  Google Scholar 

  29. Rocha-Perugini, V. et al. Tetraspanins CD9 and CD151 at the immune synapse support T cell integrin signaling. Eur. J. Immunol. 44, 1967–1975 (2014).

    CAS  Google Scholar 

  30. Toyo-Oka, K. et al. Association of a tetraspanin CD9 with CD5 on the T cell surface: role of particular transmembrane domains in the association. Int. Immunol. 11, 2043–2052 (1999).

    CAS  Google Scholar 

  31. Kerrien, S. et al. IntAct—open source resource for molecular interaction data. Nucleic Acids Res. 35, D561–D565 (2007).

    CAS  Google Scholar 

  32. Tirosh, I. et al. Dissecting the multicellular ecosystem of metastatic melanoma by single-cell RNA-seq. Science 352, 189–196 (2016).

    CAS  Google Scholar 

  33. Trickett, A. & Kwan, Y. L. T cell stimulation and expansion using anti-CD3/CD28 beads. J. Immunol. Methods 275, 251–255 (2003).

    CAS  Google Scholar 

  34. Kellogg, R. A., Tian, C., Etzrodt, M. & Tay, S. Cellular decision-making by non-integrative processing of TLR inputs. Cell Rep. 19, 125–135 (2017).

    CAS  Google Scholar 

  35. Ozinsky, A. et al. The repertoire for pattern recognition of pathogens by the innate immune system is defined by cooperation between Toll-like receptors. Proc. Natl Acad. Sci. USA 97, 13766–13771 (2000).

    CAS  Google Scholar 

  36. Qiu, Y. et al. Divergent roles of amino acid residues inside and outside the BB loop affect human Toll-like receptor (TLR)2/2, TLR2/1 and TLR2/6 responsiveness. PLoS ONE 8, e61508 (2013).

    CAS  Google Scholar 

  37. Silverstein, R. L. & Febbraio, M. CD36, a scavenger receptor involved in immunity, metabolism, angiogenesis and behavior. Sci. Signal. 2, re3 (2009).

    Google Scholar 

  38. Biedroń, R., Peruń, A. & Józefowski, S. CD36 differently regulates macrophage responses to smooth and rough lipopolysaccharide. PLoS ONE 11, e0153558 (2016).

    Google Scholar 

  39. Seimon, T. A. et al. Atherogenic lipids and lipoproteins trigger CD36–TLR2-dependent apoptosis in macrophages undergoing endoplasmic reticulum stress. Cell Metab. 12, 467–482 (2010).

    CAS  Google Scholar 

  40. Stewart, C. R. et al. CD36 ligands promote sterile inflammation through assembly of a Toll-like receptor 4 and 6 heterodimer. Nat. Immunol. 11, 155–161 (2009).

    Google Scholar 

  41. Mereu, E. et al. Benchmarking single-cell RNA-sequencing protocols for cell atlas projects. Nat. Biotechnol. 38, 747–755 (2020).

    CAS  Google Scholar 

  42. Ding, J. et al. Systematic comparison of single-cell and single-nucleus RNA-sequencing methods. Nat. Biotechnol. 38, 737–746 (2020).

    CAS  Google Scholar 

  43. Zhang, X. et al. Comparative analysis of droplet-based ultra-high-throughput single-cell RNA-seq systems. Mol. Cell 73, 130–142 (2019).

    Google Scholar 

  44. Phan, H. V. et al. High-throughput RNA sequencing of paraformaldehyde-fixed single cells. Nat. Commun. 12, 5636 (2021).

    CAS  Google Scholar 

  45. Denisenko, E. et al. Systematic assessment of tissue dissociation and storage biases in single-cell and single-nucleus RNA-seq workflows. Genome Biol. 21, 130 (2020).

    CAS  Google Scholar 

  46. van Dijk, D. et al. Recovering gene interactions from single-cell data using data diffusion. Cell 174, 716–729 (2018).

    Google Scholar 

  47. Stevens, T. J. et al. 3D structures of individual mammalian genomes studied by single-cell Hi-C. Nature 544, 59–64 (2017).

    CAS  Google Scholar 

  48. Shahi, P., Kim, S. C., Haliburton, J. R., Gartner, Z. J. & Abate, A. R. Abseq: ultrahigh-throughput single cell protein profiling with droplet microfluidic barcoding. Sci. Rep. 7, 44447 (2017).

    CAS  Google Scholar 

  49. Specht, H. et al. Single-cell proteomic and transcriptomic analysis of macrophage heterogeneity using SCoPE2. Genome Biol. 22, 50 (2021).

    CAS  Google Scholar 

  50. Zhu, Y. et al. Proteomic analysis of single mammalian cells enabled by microfluidic nanodroplet sample preparation and ultrasensitive NanoLC-MS. Angew. Chem. Int. Ed. Engl. 57, 12370–12374 (2018).

    CAS  Google Scholar 

  51. Huttlin, E. L. et al. The BioPlex Network: a systematic exploration of the human interactome. Cell 162, 425–440 (2015).

    CAS  Google Scholar 

  52. Rual, J.-F. et al. Towards a proteome-scale map of the human protein–protein interaction network. Nature 437, 1173–1178 (2005).

    CAS  Google Scholar 

  53. Grant, D. M. et al. Multiplexed FRET to image multiple signaling events in live cells. Biophys. J. 95, L69–L71 (2008).

    CAS  Google Scholar 

  54. Gong, H. et al. Simple method to prepare oligonucleotide-conjugated antibodies and its application in multiplex protein detection in single cells. Bioconjug. Chem. 27, 217–225 (2016).

    CAS  Google Scholar 

  55. Hui, E. et al. T cell co-stimulatory receptor CD28 is a primary target for PD-1-mediated inhibition. Science 355, 1428–1433 (2017).

    CAS  Google Scholar 

Download references

Acknowledgements

We thank J. Huang (Pritzker School of Molecular Imaging, University of Chicago) for the generous gift of the Jurkat and Raji cell lines used in this study. We thank A. Basu (Section of Genetic Medicine, Department of Medicine, University of Chicago) and H. Eckart (Section of Genetic Medicine, Department of Medicine, University of Chicago) for their advice on the Drop-seq protocol. We acknowledge both The University of Chicago Genomics Facility, The University of Chicago Cytometry and Antibody Technology facility and The University of Chicago Research Computing Center for their services. We thank U. Landegren (Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University) for advice with PLA. We thank Z. Ren and C. Chen at the CRI Bioinformatics Core for their advice on the alignment program. We thank A. A. Khan (Department of Pathology, University of Chicago) and D. Reiman (Department of Bioengineering, University of Illinois at Chicago) for discussions on data analysis. Automated library preparation was performed by the Cellular Screening Center at the University of Chicago. S.T. was awarded a National Institutes of Health (NIH) R01 grant GM127527 and the Paul G. Allen Distinguished Investigator Award, which supported this work. M.C. was awarded NIH R01 grants GM126553 and HG011883, and an NSF grant 2016307, which supported this work.

Author information

Authors and Affiliations

Authors

Contributions

L.V., H.V.P., C.J., S.T.R. and S.T. conceived of and designed the project. L.V., H.V.P., B.K. and C.J. performed the experiments, designed the Prox-seq probe components and analyzed sequencing data. H.V.P. and M.C. performed statistical analysis. L.V., H.V.P. and S.T. wrote the manuscript. S.T. supervised the project. All authors reviewed the manuscript.

Corresponding author

Correspondence to Savaş Tay.

Ethics declarations

Competing interests

The authors declare no competing interests.

Peer review

Peer review information

Nature Methods thanks Nikolai Slavov, Chun Ye, and the other, anonymous, reviewer for their contribution to the peer review of this work. Primary Handling Editor: Lei 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 Jurkat cell protein expression levels.

Flow cytometry data showing Prox-seq probe binding on Jurkat cells. (a) Each T cell marker in the panel along with isotype controls. (b) The gating strategy to identify individual cells. CD45RA uses the mouse IgG2a control, CD147 uses the goat control, and the rest use the mouse IgG1 control.

Extended Data Fig. 2 Raji cell protein expression levels.

Flow cytometry data showing Prox-seq probe binding on Raji cells. (a), Each B cell marker in the panel along with isotype controls. (b) The gating strategy to identify individual cells. B7 and ICAM1 use the mouse IgG1 control, HLA-DR uses the mouse IgG2a control, PDL1 uses the mouse IgG2b control, and CD147 uses the goat control.

Extended Data Fig. 3 Comparison of protein quantification between Prox-seq and flow cytometry.

(a, b) Distribution of the protein abundance of Jurkat markers as measured by (a) Prox-seq and (b) flow cytometry. (c, d) Distribution of the protein abundance of Raji markers as measured by (c) Prox-seq and (d) flow cytometry. (e) Scatter plot showing the median protein abundance as measured by flow cytometry or Prox-seq. Each point indicates a protein. The plot also shows the Spearman’s correlation coefficient, ρ, between Prox-seq and flow cytometry measurements.

Source data

Extended Data Fig. 4 Benchmarking of protein quantification based on PLA products.

(a) Schematic showing how the use of free oligo binding could help measure non-proximal Prox-seq probes. After the ligation step in the standard Prox-seq protocol, free DNA oligos were added so that they can be ligated to probes that are bound to protein but are not proximal to another Prox-seq probe. Antibodies’ cartoons were made with BioRender’s academic license. (b) Box plots showing the fraction of protein counts calculated from PLA products to the protein counts calculated from PLA products and non-proximal Prox-seq probes (n = 95 Jurkat cells and 93 Raji cells). The center line of the box indicates the median, the bottom and top bounds of the box indicate the 25th and 75th percentiles, and the whiskers extend to 1.5× the interquartile range beyond the box. (c) Scatter plots comparing protein quantification based on PLA products vs. free oligo binding method for Jurkat cell markers. (d) Scatter plots comparing protein quantification based on PLA products vs. free oligo for Raji cell markers. (e) Scatter plots comparing CD147 protein quantification based on PLA products vs. free oligo binding method for Jurkat and Raji cells. In (ce), the numbers above each panel indicate the Pearson’s correlation coefficients. The Free oligo-based estimates are made by taking the number of PLA UMI’s that contain one barcode from the indicated protein and one barcode from the free oligo.

Source data

Extended Data Fig. 5 Analysis of Prox-seq probe non-specific background binding.

(a) Schematic of the experiment. Jurkat and Raji cells were separately incubated with the full or half Prox-seq probe panel, then combined and processed with the 10x Prox-seq pipeline. The half probe panel includes Jurkat markers CD28, PD1, and CD147 probes, and Raji markers HLADR and PDL1. The full probe panel contains all the probes in the half panel, plus Jurkat marker CD3 and Raji markers ICAM1 and B7. (b) t-SNE plot based on PLA product data showing the cell type and probe panel identity. F and H stands for full and half panels, respectively (n = 856, 2738, 1159 and 1051 single cells for Jurkat_F, Jurkat_H, Raji_F and Raji_H, respectively). (c) t-SNE plots showing the expression levels of CD3E and HLA-DRA genes. (d) Plots showing the median counts of non-specific PLA products across different cell types and probe panels. The center line of the box indicates the median, the bottom and top bounds of the box indicate the 25th and 75th percentiles, and the whiskers extend to 1.5× the interquartile range beyond the box. Each black line connects the median counts of a non-specific PLA product in the half and full probe panel. n = 16 non-specific PLA products for both Jurkat and Raji clusters. (e) Violin plots showing the relative levels of Jurkat-specific PLA products CD28:CD28, PD1:PD1 and CD3:CD3, and Raji-specific PLA products HLADR:HLADR, PDL1:PDL1 and ICAM1:ICAM1. CD3:CD3 and ICAM1:ICAM1 PLA products were expected to only be detected in full probe panel clusters. (f) Heatmap showing the relative level (row z-score) of top 10 PLA product markers of each of the 4 clusters identified in (c). (g) Heatmaps showing the average log-transformed levels of protein complexes in Jurkat cells. (h) Violin plot showing the normalized levels of protein complexes CD28:CD28 and CD28:PD1 in Jurkat cells. The normalized levels were calculated by log-transforming counts per 10,000 UMIs of predicted protein complexes plus a pseudocount of 1. The fold-change was calculated by dividing the average normalized level of the Jurkat full panel cells by that of the Jurkat half panel cells.

Source data

Extended Data Fig. 6 Characterization of CD4 T cells in PBMCs using plate-based Prox-seq.

(a) Scatter plot showing two subpopulations of CD4 T cells, according to CD9-related PLA products level. (b) Violin plot showing that, unlike CD8 T cells, both subpopulations of CD4 T cells do not express the protein complex CD9:CD8. (c) Violin plots showing the distribution of proteins CD3, CD4 and CD9 in the two subpopulations of CD4 T cells. Note that the complex detection algorithm assigns zero values to low-abundance PLA products that do not pass the statistical test.

Source data

Extended Data Fig. 7 Number of predicted protein complexes across cell types.

(a) t-SNE plot of PBMC clusters, obtained using mRNA expression level. (b) Violin plots showing the number of predicted protein complexes per single cell, for each of the 8 clusters identified using mRNA data. The horizontal red lines indicate the total number of predicted protein complexes per cluster. In total, 61 protein complexes were detected across all 8 clusters, of which 37 complexes are unique. (c) Plot showing the number of protein complexes predicted by the algorithm at different number of cells. Here, various numbers of cells were randomly subsampled from each of the 8 clusters identified in (a), and the complex prediction algorithm was applied on the subsampled cells.

Source data

Extended Data Fig. 8 Analysis of LPS and PAM-treated macrophages.

(ac) Receiver operating characteristic (ROC) curves of 5-fold cross-validation of a logistic regression classifier that is trained on (a) 5-minute data, (b) 2-hour data, and (c) 12-hour data. The black dashed lines in (ac) indicate random classification. (d) Violin plots showing the log-transformed count of the top three PLA products of the logistic regression model that is trained on 2-hour data. P-values are calculated using two-sided Welch’s t-test (n = 31, 32, 32, and 36 single cells for the control, LPS, PAM, and both treatment groups, respectively). (e) Schematic showing how the logistic regression classifier is used to predict response (LPS-like, PAM-liked, and mixed) in cells treated with both LPS and PAM after 2 h. (f) Bar plot showing the proportion of LPS/PAM-treated cells that show LPS-like, PAM-like and mixed response. n indicates the number of cells in each response group. (g) Violin plots showing the log-transformed count of the top logistic regression coefficients (Fig. 6b) in each predicted response group for cells treated with both ligands. (h, i) Heatmaps showing (h) the relative PLA product levels and (i) the relative protein levels of the LPS-like, PAM-like and mixed response groups. The PLA product (or protein) counts are log-transformed, then averaged by response group, and finally standardized. Hierarchical clustering is performed on the PLA products (or proteins) and response groups using Euclidean distance and complete linkage. (j) ROC curves of a logistic regression classifier trained on protein levels from different time points. The classifier is trained to predict whether a single cell was stimulated with LPS or PAM. Each ROC curve represents the mean ROC curve from 5-fold cross-validation. The area under the curve (AUC) metric of each time point is presented as mean ± s.d. of the AUC metrics from 5-fold cross-validation for that particular time point.

Source data

Extended Data Fig. 9 Analysis of sequencing depth.

(ac) Effects of sequencing depth on (a) the number of detected genes and transcript counts per single cell, (b) the number of detected PLA products and their UMI counts, and (c) the number of detected proteins and protein UMI counts in 10x-based Prox-seq. (d) Effects of sequencing depth on automated cell type annotation based on mRNA data with singleR package. The cell type annotation at the maximum sequencing depth is used as the ground truth annotation. (e) Effects of sequencing depth on the number of detected protein complexes. Clusters were identified using mRNA data (see Extended Data Fig. 7). Clusters 0 and 3 were chosen as examples because they had the most number of cells per cluster. In (ae), the sequencing results of the mRNA and PLA product libraries from the 10x PBMC experiment were downsampled to 10%, 20%, 40%, 60%, and 80% to simulate different sequencing depths. (f, g) Effects of sequencing depth on (f) the number of detected PLA products and their UMI counts, and (g) the number of detected proteins and protein UMI counts in plate-based Prox-seq. In (f, g), the sequencing results of the mRNA and PLA product libraries from the plate-based PBMC experiment were downsampled to 0.5%, 1%, 5%, 10%, 25%, 50%, and 75% to simulate different sequencing depths. The red dashed lines in (e, f) indicate 10,000 mean reads per cell.

Source data

Supplementary information

Supplementary information

Supplementary Figs. 1–11, captions for Supplementary Tables 1–12 and Supplementary Methods and references.

Reporting Summary

Supplementary Tables 1–12

Supplementary tables 1–12

Supplementary Data 1

List of identified protein complexes.

Supplementary Data 2

List of gene expression levels.

Source data

Source Data

Statistical source data for Figs. 1–6, Extended Data Figs. 3–9 and Supplementary Figs. 2–11.

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

Vistain, L., Van Phan, H., Keisham, B. et al. Quantification of extracellular proteins, protein complexes and mRNAs in single cells by proximity sequencing. Nat Methods 19, 1578–1589 (2022). https://doi.org/10.1038/s41592-022-01684-z

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41592-022-01684-z

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