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:

Time-resolved assessment of single-cell protein secretion by sequencing

Abstract

Secreted proteins play critical roles in cellular communication. Methods enabling concurrent measurement of cellular protein secretion, phenotypes and transcriptomes are still unavailable. Here we describe time-resolved assessment of protein secretion from single cells by sequencing (TRAPS-seq). Released proteins are trapped onto the cell surface and probed by oligonucleotide-barcoded antibodies before being simultaneously sequenced with transcriptomes in single cells. We demonstrate that TRAPS-seq helps unravel the phenotypic and transcriptional determinants of the secretion of pleiotropic TH1 cytokines (IFNγ, IL-2 and TNF) in activated T cells. In addition, we show that TRAPS-seq can be used to track the secretion of multiple cytokines over time, uncovering unique molecular signatures that govern the dynamics of single-cell cytokine secretions. Our results revealed that early central memory T cells with CD45RA expression (TCMRA) are important in both the production and maintenance of polyfunctional cytokines. TRAPS-seq presents a unique tool for seamless integration of secretomics measurements with multi-omics profiling in single cells.

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: Schematic representation of TRAPS-seq workflow.
Fig. 2: TRAPS-seq enables integrated profiling of single-cell protein secretion, phenotypes and transcriptomes.
Fig. 3: TRAPS-seq identifies TCMRA cells as the most effective multiple-cytokine-secreting T cells.
Fig. 4: Differential gene signatures associated with cytokine secretion pattern.
Fig. 5: Secretion tracing reveals secretion strength-associated acquisition and maintenance of cytokine-releasing polyfunctionality.
Fig. 6: Secretion tracing with TRAPS-seq identifies cellular and molecular determinants of cytokine secretion dynamics.

Similar content being viewed by others

Data availability

Curated hallmark gene sets are publicly available from MSigDB (v7). Single-cell gene expression, surface marker and secreted cytokine abundance data have been deposited to the Gene Expression Omnibus (GEO) under accession number GSE200690. Source data are provided with this paper.

Code availability

The R code to reproduce the analyses in the figures is available from the Zenodo repository (https://doi.org/10.5281/zenodo.7515637).

References

  1. Kumar, B. V., Connors, T. J. & Farber, D. L. Human T cell development, localization, and function throughout life. Immunity 48, 202–213 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Grivennikov, S. I., Greten, F. R. & Karin, M. Immunity, inflammation, and cancer. Cell 140, 883–899 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Dranoff, G. Cytokines in cancer pathogenesis and cancer therapy. Nat. Rev. Cancer 4, 11–22 (2004).

    Article  CAS  PubMed  Google Scholar 

  4. Uhlén, M. et al. The human secretome. Sci. Signal. 12, eaaz0274 (2019).

    Article  PubMed  Google Scholar 

  5. Antonioli, L., Blandizzi, C., Pacher, P., Guilliams, M. & Haskó, G. Rethinking communication in the immune system: the quorum sensing concept. Trends Immunol. 40, 88–97 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Appay, V., van Lier, R. A. W., Sallusto, F. & Roederer, M. Phenotype and function of human T lymphocyte subsets: consensus and issues. Cytometry A 73, 975–983 (2008).

    Article  PubMed  Google Scholar 

  7. Varadarajan, N. et al. A high-throughput single-cell analysis of human CD8+ T cell functions reveals discordance for cytokine secretion and cytolysis. J. Clin. Invest. 121, 4322–4331 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Favre, N., Bordmann, G. & Rudin, W. Comparison of cytokine measurements using ELISA, ELISPOT and semi-quantitative RT-PCR. J. Immunol. Methods 204, 57–66 (1997).

    Article  CAS  PubMed  Google Scholar 

  9. Manz, R., Assenmacher, M., Pflüger, E., Miltenyi, S. & Radbruch, A. Analysis and sorting of live cells according to secreted molecules, relocated to a cell-surface affinity matrix. Proc. Natl Acad. Sci. USA 92, 1921–1925 (1995).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Burel, J. G., Apte, S. H., Groves, P. L., McCarthy, J. S. & Doolan, D. L. Polyfunctional and IFN-γ monofunctional human CD4+ T cell populations are molecularly distinct. JCI Insight 2, e87499 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  11. Pinder, C. L. et al. Isolation and characterization of antigen-specific plasmablasts using a novel flow cytometry-based Ig capture assay. J. Immunol. 199, 4180–4188 (2017).

    Article  CAS  PubMed  Google Scholar 

  12. Love, J. C., Ronan, J. L., Grotenbreg, G. M., van der Veen, A. G. & Ploegh, H. L. A microengraving method for rapid selection of single cells producing antigen-specific antibodies. Nat. Biotechnol. 24, 703–707 (2006).

    Article  CAS  PubMed  Google Scholar 

  13. Han, Q. et al. Polyfunctional responses by human T cells result from sequential release of cytokines. Proc. Natl Acad. Sci. USA 109, 1607–1612 (2012).

    Article  CAS  PubMed  Google Scholar 

  14. Lu, Y. et al. Highly multiplexed profiling of single-cell effector functions reveals deep functional heterogeneity in response to pathogenic ligands. Proc. Natl Acad. Sci. USA 112, E607–E615 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Stanley, A. C. & Lacy, P. Pathways for cytokine secretion. Physiology 25, 218–229 (2010).

    Article  CAS  PubMed  Google Scholar 

  16. de Saint Basile, G., Ménasché, G. & Fischer, A. Molecular mechanisms of biogenesis and exocytosis of cytotoxic granules. Nat. Rev. Immunol. 10, 568–579 (2010).

    Article  Google Scholar 

  17. Zheng, Y., Tang, L., Mabardi, L., Kumari, S. & Irvine, D. J. Enhancing adoptive cell therapy of cancer through targeted delivery of small-molecule immunomodulators to internalizing or noninternalizing receptors. ACS Nano 11, 3089–3100 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Friedl, P. & Gunzer, M. Interaction of T cells with APCs: the serial encounter model. Trends Immunol. 22, 187–191 (2001).

    Article  CAS  PubMed  Google Scholar 

  19. Stoeckius, M. et al. Cell Hashing with barcoded antibodies enables multiplexing and doublet detection for single cell genomics. Genome Biol. 19, 224 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Nicolet, B. P. & Wolkers, M. C. The relationship of mRNA with protein expression in CD8+ T cells associates with gene class and gene characteristics. PLoS ONE 17, e0276294 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Garcia-Sanz, J. A. & Lenig, D. Translational control of interleukin 2 messenger RNA as a molecular mechanism of T cell anergy. J. Exp. Med. 184, 159–164 (1996).

    Article  CAS  PubMed  Google Scholar 

  23. Villarino, A. V. et al. Posttranscriptional silencing of effector cytokine mRNA underlies the anergic phenotype of self-reactive T cells. Immunity 34, 50–60 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Salerno, F., Paolini, N. A., Stark, R., von Lindern, M. & Wolkers, M. C. Distinct PKC-mediated posttranscriptional events set cytokine production kinetics in CD8+ T cells. Proc. Natl Acad. Sci. USA 114, 9677–9682 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Gattinoni, L. et al. Acquisition of full effector function in vitro paradoxically impairs the in vivo antitumor efficacy of adoptively transferred CD8+ T cells. J. Clin. Invest. 115, 1616–1626 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. He, X.-S. et al. T cell–dependent production of IFN-γ by NK cells in response to influenza A virus. J. Clin. Invest. 114, 1812–1819 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Seder, R. A., Darrah, P. A. & Roederer, M. T-cell quality in memory and protection: implications for vaccine design. Nat. Rev. Immunol. 8, 247–258 (2008).

    Article  CAS  PubMed  Google Scholar 

  28. Panagioti, E., Klenerman, P., Lee, L. N., van der Burg, S. H. & Arens, R. Features of effective T cell-inducing vaccines against chronic viral infections. Front. Immunol. 9, 276 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  29. Rossi, J. et al. Preinfusion polyfunctional anti-CD19 chimeric antigen receptor T cells are associated with clinical outcomes in NHL. Blood 132, 804–814 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Wu, T., Luah, Y. H., Luo, Y., Womersley, H. J. & Cheow, L. F. Label-free lymphocytes reconstitution using side scatter for optimal T cell manufacturing. Preprint at bioRxiv https://doi.org/10.1101/2020.11.09.375584 (2020).

  31. Fuertes Marraco, S. A. et al. Long-lasting stem cell–like memory CD8+ T cells with a naïve-like profile upon yellow fever vaccination. Sci. Transl. Med. 7, 282ra48 (2015).

    Article  PubMed  Google Scholar 

  32. van den Broek, T., Borghans, J. A. M. & van Wijk, F. The full spectrum of human naive T cells. Nat. Rev. Immunol. 18, 363–373 (2018).

    Article  PubMed  Google Scholar 

  33. Moroz, A. et al. IL-21 enhances and sustains CD8+ T cell responses to achieve durable tumor immunity: comparative evaluation of IL-2, IL-15, and IL-21. J. Immunol. 173, 900–909 (2004).

    Article  CAS  PubMed  Google Scholar 

  34. Upadhyay, V. & Fu, Y.-X. Lymphotoxin signalling in immune homeostasis and the control of microorganisms. Nat. Rev. Immunol. 13, 270–279 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Rygiel, T. P. et al. Lack of CD200 enhances pathological T cell responses during influenza infection. J. Immunol. 183, 1990–1996 (2009).

    Article  CAS  PubMed  Google Scholar 

  36. Kotwica-Mojzych, K., Jodłowska-Jędrych, B. & Mojzych, M. CD200:CD200R interactions and their importance in immunoregulation. Int. J. Mol. Sci. 22, 1602 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Chung, J.-S., Dougherty, I., Cruz, P. D.Jr. & Ariizumi, K. Syndecan-4 mediates the coinhibitory function of DC-HIL on T cell activation. J. Immunol. 179, 5778–5784 (2007).

    Article  CAS  PubMed  Google Scholar 

  38. Li, J., He, Y., Hao, J., Ni, L. & Dong, C. High levels of Eomes promote exhaustion of anti-tumor CD8+ T cells. Front. Immunol. 9, 2981 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Lee, W., Kim, H. S., Hwang, S. S. & Lee, G. R. The transcription factor Batf3 inhibits the differentiation of regulatory T cells in the periphery. Exp. Mol. Med. 49, e393 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Somerville, T. D. D. et al. ZBED2 is an antagonist of interferon regulatory factor 1 and modifies cell identity in pancreatic cancer. Proc. Natl Acad. Sci. USA 117, 11471–11482 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Giang, S. & La Cava, A. IRF1 and BATF: key drivers of type 1 regulatory T-cell differentiation. Cell. Mol. Immunol. 14, 652–654 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  42. Westerhof, L. M. et al. Multifunctional cytokine production reveals functional superiority of memory CD4 T cells. Eur. J. Immunol. 49, 2019–2029 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Gierahn, T. M. et al. Seq-Well: portable, low-cost RNA sequencing of single cells at high throughput. Nat. Methods 14, 395–398 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Deng, N. & Mosmann, T. R. Optimization of the cytokine secretion assay for human IL-2 in single and combination assays. Cytometry A 87, 777–783 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Bounab, Y. et al. Dynamic single-cell phenotyping of immune cells using the microfluidic platform DropMap. Nat. Protoc. 15, 2920–2955 (2020).

    Article  CAS  PubMed  Google Scholar 

  46. Snyder, J. E. et al. Measuring the frequency of mouse and human cytotoxic T cells by the Lysispot assay: independent regulation of cytokine secretion and short-term killing. Nat. Med. 9, 231–236 (2003).

    Article  CAS  PubMed  Google Scholar 

  47. Yeh, J.-H., Sidhu, S. S. & Chan, A. C. Regulation of a late phase of T cell polarity and effector functions by Crtam. Cell 132, 846–859 (2008).

    Article  CAS  PubMed  Google Scholar 

  48. Takeuchi, A. et al. CRTAM confers late-stage activation of CD8+ T cells to regulate retention within lymph node. J. Immunol. 183, 4220–4228 (2009).

    Article  CAS  PubMed  Google Scholar 

  49. Hwang, J. et al. The structural basis for the negative regulation of thioredoxin by thioredoxin-interacting protein. Nat. Commun. 5, 2958 (2014).

    Article  PubMed  Google Scholar 

  50. Muri, J., Thut, H. & Kopf, M. The thioredoxin-1 inhibitor Txnip restrains effector T-cell and germinal center B-cell expansion. Eur. J. Immunol. 51, 115–124 (2021).

    Article  CAS  PubMed  Google Scholar 

  51. Vairapandi, M., Balliet, A. G., Hoffman, B. & Liebermann, D. A. GADD45b and GADD45g are cdc2/cyclinB1 kinase inhibitors with a role in S and G2/M cell cycle checkpoints induced by genotoxic stress. J. Cell. Physiol. 192, 327–338 (2002).

    Article  CAS  PubMed  Google Scholar 

  52. Lu, B., Ferrandino, A. F. & Flavell, R. A. Gadd45β is important for perpetuating cognate and inflammatory signals in T cells. Nat. Immunol. 5, 38–44 (2004).

    Article  CAS  PubMed  Google Scholar 

  53. Ju, S. et al. Gadd45b and Gadd45g are important for anti-tumor immune responses. Eur. J. Immunol. 39, 3010–3018 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Chatterjee, S. et al. Targeting PIM kinase with PD1 inhibition improves immunotherapeutic antitumor T-cell response. Clin. Cancer Res. 25, 1036–1049 (2019).

    Article  CAS  PubMed  Google Scholar 

  55. Daenthanasanmak, A. et al. PIM-2 protein kinase negatively regulates T cell responses in transplantation and tumor immunity. J. Clin. Invest. 128, 2787–2801 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  56. Becher, B., Tugues, S. & Greter, M. GM-CSF: from growth factor to central mediator of tissue inflammation. Immunity 45, 963–973 (2016).

    Article  CAS  PubMed  Google Scholar 

  57. Mahnke, Y. D., Brodie, T. M., Sallusto, F., Roederer, M. & Lugli, E. The who’s who of T-cell differentiation: human memory T-cell subsets. Eur. J. Immunol. 43, 2797–2809 (2013).

    Article  CAS  PubMed  Google Scholar 

  58. Croft, M., So, T., Duan, W. & Soroosh, P. The significance of OX40 and OX40L to T-cell biology and immune disease. Immunol. Rev. 229, 173–191 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Mahajan, S. et al. The role of ICOS in the development of CD4 T cell help and the reactivation of memory T cells. Eur. J. Immunol. 37, 1796–1808 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Trambas, C. M. & Griffiths, G. M. Delivering the kiss of death. Nat. Immunol. 4, 399–403 (2003).

    Article  CAS  PubMed  Google Scholar 

  61. Enesa, K. et al. Hydrogen peroxide prolongs nuclear localization of NF-κB in activated cells by suppressing negative regulatory mechanisms. J. Biol. Chem. 283, 18582–18590 (2008).

    Article  CAS  PubMed  Google Scholar 

  62. Caceres, P. S., Gravotta, D., Zager, P. J., Dephoure, N. & Rodriguez-Boulan, E. Quantitative proteomics of MDCK cells identify unrecognized roles of clathrin adaptor AP-1 in polarized distribution of surface proteins. Proc. Natl Acad. Sci. USA 116, 11796–11805 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. La Manno, G. et al. RNA velocity of single cells. Nature 560, 494–498 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  64. Qiu, Q. et al. Massively parallel and time-resolved RNA sequencing in single cells with scNT-seq. Nat. Methods 17, 991–1001 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Achar, S. R. et al. Universal antigen encoding of T cell activation from high-dimensional cytokine dynamics. Science 376, 880–884 (2022).

    Article  CAS  PubMed  Google Scholar 

  66. Bhattacharyya, N. D. et al. TCR affinity controls the dynamics but not the functional specification of the antimycobacterial CD4+ T cell response. J. Immunol. 206, 2875–2887 (2021).

    Article  CAS  PubMed  Google Scholar 

  67. Roelli, P., Flynn, B. & Gui, G. Hoohm/CITE-seq-Count: 1.4.2 (Version 1.4.2). Zenodo https://doi.org/10.5281/zenodo.2590196 (2019).

  68. Hao, Y. et al. Integrated analysis of multimodal single-cell data. Cell 184, 3573–3587.e29 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  69. Hafemeister, C. & Satija, R. Normalization and variance stabilization of single-cell RNA-seq data using regularized negative binomial regression. Genome Biol. 20, 296 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  70. Yu, G., Wang, L.-G., Han, Y. & He, Q.-Y. clusterProfiler: an R package for comparing biological themes among gene clusters. OMICS 16, 284–287 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  71. Subramanian, A. et al. Gene set enrichment analysis: a knowledge-based approach for interpreting genome-wide expression profiles. Proc. Natl Acad. Sci. USA 102, 15545–15550 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  72. Makowski, D., Ben-Shachar, M. S., Patil, I. & Lüdecke, D. Methods and algorithms for correlation analysis in R. J. Open Source Softw. 5, 2306 (2020).

    Article  Google Scholar 

Download references

Acknowledgements

We thank M. Birnbaum (MIT) and J.Y.-H. Loh (A*STAR) for the generous gifts of Raji and Jurkat cell lines, respectively, used in this study. We acknowledge technical support from the Flow Cytometry Laboratory (NUS Medical Sciences Cluster) for cell sorting and thank K.C. Tang from iHealthtech for assistance in the generation of silicon mold. This work was supported by funding from the National Medical Research Council Individual Research Grant (MOH-000219), Singapore Ministry of Education Academic Research Fund Tier 2 (MOE-000063) and Institute for Health Innovation and Technology (iHealthtech), NUS.

Author information

Authors and Affiliations

Authors

Contributions

T.W. and L.F.C. conceived and designed the study. T.W. performed the experiments and data analysis, with input from L.F.C. and J.S. in data analysis and assistance from H.J.W. and J.R.W. in sequencing. T.W. and L.F.C. wrote the paper with feedback from J.S. All authors approved the submission.

Corresponding author

Correspondence to Lih Feng Cheow.

Ethics declarations

Competing interests

L.F.C. and T.W. are listed as co-inventors on a patent application related to this work (patent application no. PCT/SG2022/050812). The other authors declare no competing interests.

Peer review

Peer review information

Nature Methods thanks Xiangjie Li and the other, anonymous, reviewers for their contribution to the peer review of this work. Primary Handling Editor: Madhura Mukhopadhyay, in collaboration with the Nature Methods team. Peer reviewer reports are available.

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 Characterization of the accuracy of cytokine secretion capture assay.

(A) CapAb-modified PBMC (concurrently for 3 cytokines) were pulsed with saturated concentrations of purified IFN-γ, IL-2 and TNF-α (200 ng/ml each), or subjected to secretion capture assay after 6-hour activation with 50 ng/ml PMA and 1 µg/ml ionomycin. Flow cytometry data shows that CapAb cytokine binding sites are not saturated under physiological secretion conditions. (B, C) Pre-stimulated PBMC (6 hours) and unstimulated Jurkat cells were modified with CapAb and mixed together before subjected to secretion capture measurements (B). Cells with significant cytokine secretion can be found within the stimulated CD3+ lympbocyte population. Instead, negligible cytokine capture was observed on the unstimulated Jurkat cells in this mixture experiment (C). (D) The percentage of cytokine-secreting cells among CD3+ lymphocytes are comparable with or without Jurkat cells added. Data are mean ± S.E.M of three independent experiments.

Source data

Extended Data Fig. 2 Cell clustering and annotation.

(AC) Unsupervised cell clustering based on the expression of a panel of surface protein markers as listed in B. The ‘negative population’ (blue arrow) was determined as the cluster with the lowest specific protein expression. Shown are UMAP plots for all cells from integrated time points (A) as well as for cells from individual time points (C). (D) Manual cell type annotation based on the expression profile of surface protein markers and a panel of genes relating to immune cell activation/differentiation. (E) Cells from all time points were integrated and clustered by their gene expression profiles (left) and colored by cell subtypes (right) as annotated in D.

Source data

Extended Data Fig. 3 Refined T-cell clustering and annotation.

(A) T cells from Fig. 2F (left) were re-clustered by their surface marker expression (cell numbers for each new T-cell cluster indicated in bracket, middle). For comparison, the new UMAP plot was colored by the original cell annotation (right). (B) The new T-cell clusters were annotated according to surface protein expression and a panel of selected genes associated with T-cell activation/differentiation. (C) Sankey diagram showing the correspondence between the annotations of the original and new clustering.

Source data

Extended Data Fig. 4 Characterization of cell types associated with cytokine secretion pattern over time.

The cytokine secretion patterns of different T-cell subpopulations were shown as Sankey plots (left panel) and heatmap matrices (right panel) for time point of 1 hour, 6 hours, and 16 hours respectively post stimulation. T-cell clusters with less than 40 cells were omitted.

Source data

Extended Data Fig. 5 Measurement of single-cell cytokine secretion in microengraved arrays.

(A, B) Celltrace Far Red-labelled T cells were loaded into the nanowell array at 20,000 cells (A) or 40,000 cells (B). The observed cell occupancies in wells from two independent experiments were presented (A, B). (C) Naïve/memory composition of resting (from resting PBMC) or effector (from extensively expanded T cells) T cells that were used for the testing of on-array SCA. (D) The cells were pre-stimulated with 50 ng/ml PMA and 1 µg/ml ionomycin (‘P + I’) for 3 hours before CapAbs labelling. SCA was carried out in both tubes and arrays as described in Methods. (E) Side-by-side comparison between tube- and array-based SCA using T-cell samples shown in (C). (F) Summary of cytokine capture efficiency using tube- and array-based SCA. Shown were percentage of cells identified to be secreting cytokines and relative intensity of positive signal versus background staining (signal-to-noise ratio). Data are mean of three (array-based) and two (tube-based) independent experiments with individual data points presented.

Source data

Extended Data Fig. 6 Scatter plots of T-cell secretion-transition pattern over time.

Shown were 6-color FACS analyses for the measurement of three cytokines simultaneously over two time periods, relating to Fig. 5. A total of four secretion windows (0, 1, 6, and 16 hour) with two consecutive secretion capture each were performed to track the secretion dynamics of single activated T cells. After the 1st-round of SCA, cells were stained in-situ with a first-set of fluorescent antibodies against IFN-γ, IL-2 and TNF-α to label cytokines secreted during this first period. After buffer exchange and supplementation with additional CapAbs, the cells were further incubated in the sealed nanowells for additional 45 minutes, and stained in situ with a second set of differently-labeled fluorescent antibodies to detect cytokines secreted during the second period.

Source data

Extended Data Fig. 7 Characterization of secretory transition states.

(A) Cell clustering based on single cell transcriptome in secretion tracing experiment with cell numbers each cluster indicated in bracket. (B) Identification of non-T cells (clusters 8 and 11) according to the positive expression of non-T cell-related genes including CD19, MS4A1 and CST3 (encoding CD19, CD20 and cystatin C respectively) but absent expression of other T cell-associated genes. (C) Unsupervised T-cell clustering based on cytokine secretion dynamics derived from two consecutive rounds of secretion capture. The cells initially without any secretion detected were omitted. (D) Cell cluster annotation according to their secretion transition states.

Source data

Extended Data Fig. 8 Characterization of dynamic cytokine state transitions associated with T-cell subtypes.

(A, B) Unsupervised T-cell clustering by surface marker expression with cell numbers each cluster bracketed (A) and annotated according to the expression profile of both cell surface proteins and selected genes associated with T-cell activation/differentiation. (C) UMAP plot as shown in A was colored by secretion transitions in Extended Data Fig. 7C. (D) Distribution of T-cell subpopulations among different secretion transitions.

Source data

Extended Data Fig. 9 Concurrent measurement of the release of cytokine and cytotoxic molecule by SCA.

(A) HyNic-4FB chemistry-based covalent conjugation of anti-CD45 and anti-granzyme B (GrB) to form bispecific antibody. (B) T cells modified with CD45-GrB bispecific antibodies are able to capture extracellular GrB molecules with high efficiency. (C, D) Secretion capture of GrB, IFN-γ and TNF-α over a period of 10 hours in CD8+ T cells (C) and CD8 T cells (D) after stimulation with anti-CD3/CD28 Dynabeads (beads-to-cell ratio = 1:1) in 96-well U-bottom plate.

Source data

Extended Data Fig. 10 Characterization of CD45-independent secretion capture assay in non-immune cells.

(A) Schematic diagram shows the principle of cell-surface biotinylation with Sulfo-NHS-biotin as a universal method to create anchor sites for capture antibodies on cell surfaces when a specific surface marker is not available. Biotinylated cell surfaces can be modified with streptavidin to immobilize biotin-conjugated capture antibodies of any kind. (B) The density of capture antibody binding sites on modified HeLa (cervical epithelial cancer) and A549 cells (lung epithelial cancer) can be tuned by varying the concentrations of Sulfo-NHS-biotin as detected by Streptavidin-PE. (C) Optimization of streptavidin amount needed to maximize the binding of biotinylated capture antibodies on cell surface. (D) IL-8 secretion capture matrices were shown to successfully capture purified IL-8 in staining buffer. (E) IL-8 intracellular cytokine staining for HeLa and A549 cells that were stimulated with 5 ng/ml IL-1β for 4 hours, with the presence of Brefeldin A during the last hour. (F) IL-8 secretion capture assay using HeLa and A549 cells that were pre-stimulated with 5 ng/ml IL-1β for 4 hours. Data are mean ± S.E.M of three independent experiments, two-tailed t-test (E, F).

Source data

Supplementary information

Supplementary Information

Supplementary Methods, Figs. 1–10 and Tables 1 and 2.

Reporting Summary

Peer Review File

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 Fig. 6

Statistical source data.

Source Data Extended Data Fig. 1

Statistical source data.

Source Data Extended Data Fig. 2

Statistical source data.

Source Data Extended Data Fig. 3

Statistical source data.

Source Data Extended Data Fig. 4

Statistical source data.

Source Data Extended Data Fig. 5

Statistical source data.

Source Data Extended Data Fig. 6

Statistical source data.

Source Data Extended Data Fig. 7

Statistical source data.

Source Data Extended Data Fig. 8

Statistical source data.

Source Data Extended Data Fig. 9

Statistical source data.

Source Data Extended Data Fig. 10

Statistical source data.

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

Wu, T., Womersley, H.J., Wang, J.R. et al. Time-resolved assessment of single-cell protein secretion by sequencing. Nat Methods 20, 723–734 (2023). https://doi.org/10.1038/s41592-023-01841-y

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41592-023-01841-y

Search

Quick links

Nature Briefing: Translational Research

Sign up for the Nature Briefing: Translational Research newsletter — top stories in biotechnology, drug discovery and pharma.

Get what matters in translational research, free to your inbox weekly. Sign up for Nature Briefing: Translational Research