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.

  • Brief Communication
  • Published:

NEAT-seq: simultaneous profiling of intra-nuclear proteins, chromatin accessibility and gene expression in single cells

Abstract

In this work, we describe NEAT-seq (sequencing of nuclear protein epitope abundance, chromatin accessibility and the transcriptome in single cells), enabling interrogation of regulatory mechanisms spanning the central dogma. We apply this technique to profile CD4 memory T cells using a panel of master transcription factors (TFs) that drive T cell subsets and identify examples of TFs with regulatory activity gated by transcription, translation and regulation of chromatin binding. We also link a noncoding genome-wide association study single-nucleotide polymorphism (SNP) within a GATA motif to a putative target gene, using NEAT-seq data to internally validate SNP impact on GATA3 regulation.

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: An intra-nuclear staining protocol using oligo-antibodies enables simultaneous profiling of nuclear proteins, chromatin accessibility and RNA transcripts in single cells.
Fig. 2: Profiling of CD4 memory T cells using NEAT-seq reveals modes of regulation across the central dogma.
Fig. 3: Identification of peak–gene linkages associated with master TF protein expression.

Similar content being viewed by others

Data availability

Raw and processed sequencing data generated in this study are available through GEO (GSE178707). Published bone marrow and peripheral blood single-cell ATAC-seq and RNA-seq data were obtained from GSE139369. The CISBP database is available at http://cisbp.ccbr.utoronto.ca/. The Transfac database is available at https://genexplain.com/transfac/. Source data are provided with this paper.

Code availability

Code used for analysis and figures are available at https://github.com/GreenleafLab/NEAT-seq_reproducibility.

References

  1. Ma, S. et al. Chromatin potential identified by shared single-cell profiling of RNA and chromatin. Cell 183, 1103–1116.e20 (2020).

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

  3. Swanson, E. et al. Simultaneous trimodal single-cell measurement of transcripts, epitopes, and chromatin accessibility using TEA-seq. eLife https://doi.org/10.7554/eLife.63632 (2021).

  4. Mimitou, E. P. et al. Scalable, multimodal profiling of chromatin accessibility, gene expression and protein levels in single cells. Nat. Biotechnol. https://doi.org/10.1038/s41587-021-00927-2 (2021).

  5. Gerlach, J. P. et al. Combined quantification of intracellular (phospho-)proteins and transcriptomics from fixed single cells. Sci. Rep. 9, 1469 (2019).

    Article  Google Scholar 

  6. Reimegård, J. et al. A combined approach for single-cell mRNA and intracellular protein expression analysis. Commun. Biol. 4, 624 (2021).

    Article  Google Scholar 

  7. Rivello, F. et al. Single-cell intracellular epitope and transcript detection revealing signal transduction dynamics. Cell. Rep. Meth. 1, 100070 (2021).

  8. Chung, H. et al. Joint single-cell measurements of nuclear proteins and RNA in vivo. Nat. Methods 18, 1204–1212 (2021).

    Article  CAS  Google Scholar 

  9. Spitz, F. & Furlong, E. E. M. Transcription factors: from enhancer binding to developmental control. Nat. Rev. Genet. 13, 613–626 (2012).

    Article  CAS  Google Scholar 

  10. Grün, D., Kester, L. & van Oudenaarden, A. Validation of noise models for single-cell transcriptomics. Nat. Methods 11, 637–640 (2014).

    Article  Google Scholar 

  11. Marinov, G. K. et al. From single-cell to cell-pool transcriptomes: stochasticity in gene expression and RNA splicing. Genome Res. 24, 496–510 (2014).

    Article  CAS  Google Scholar 

  12. Gillespie, M. A. et al. Absolute quantification of transcription factors reveals principles of gene regulation in erythropoiesis. Mol. Cell 78, 960–974.e11 (2020).

    Article  CAS  Google Scholar 

  13. Mimitou, E. P. et al. Multiplexed detection of proteins, transcriptomes, clonotypes and CRISPR perturbations in single cells. Nat. Methods 16, 409–412 (2019).

    Article  CAS  Google Scholar 

  14. 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  Google Scholar 

  15. Wang, Y. et al. Multiplexed in situ protein imaging using DNA-barcoded antibodies with extended hybridization chain reactions. Preprint at bioRxiv https://doi.org/10.1101/274456 (2021).

  16. Sallusto, F. & Lanzavecchia, A. Heterogeneity of CD4+ memory T cells: functional modules for tailored immunity. Eur. J. Immunol. 39, 2076–2082 (2009).

    Article  CAS  Google Scholar 

  17. Fang, D. & Zhu, J. Dynamic balance between master transcription factors determines the fates and functions of CD4 T cell and innate lymphoid cell subsets. J. Exp. Med. 214, 1861–1876 (2017).

    Article  CAS  Google Scholar 

  18. Barski, A. et al. Rapid recall ability of memory T cells is encoded in their epigenome. Sci. Rep. 7, 39785 (2017).

    Article  CAS  Google Scholar 

  19. Samstein, R. M. et al. Foxp3 exploits a pre-existent enhancer landscape for regulatory T cell lineage specification. Cell 151, 153–166 (2012).

    Article  CAS  Google Scholar 

  20. Cova, G. et al. Helios represses megakaryocyte priming in hematopoietic stem and progenitor cells. J. Exp. Med. 218, e20202317 (2021).

  21. Sun, G. D. et al. The endoplasmic reticulum stress-inducible protein Niban regulates eIF2alpha and S6K1/4E-BP1 phosphorylation. Biochem. Biophys. Res. Commun. 360, 181–187 (2007).

    Article  CAS  Google Scholar 

  22. Cook, K. D. & Miller, J. TCR-dependent translational control of GATA-3 enhances Th2 differentiation. J. Immunol. 185, 3209–3216 (2010).

    Article  CAS  Google Scholar 

  23. Sonenberg, N. & Hinnebusch, A. G. Regulation of translation initiation in eukaryotes: mechanisms and biological targets. Cell 136, 731–745 (2009).

  24. Schmiedel, B. J. et al. Impact of genetic polymorphisms on human immune cell gene expression. Cell 175, 1701–1715.e16 (2018).

    Article  CAS  Google Scholar 

  25. Taylor, K. E., Mark Ansel, K., Marson, A., Criswell, L. A. & Farh, K. K.-H. PICS2: next-generation fine mapping via probabilistic identification of causal SNPs. Bioinformatics https://doi.org/10.1093/bioinformatics/btab122 (2021).

  26. Alkes Group, UKBB summary statistics, https://alkesgroup.broadinstitute.org/ (2020).

  27. Gieseck, R. L., Wilson, M. S. & Wynn, T. A. Type 2 immunity in tissue repair and fibrosis. Nat. Rev. Immunol. 18, 62–76 (2018).

    Article  CAS  Google Scholar 

  28. Peters, M. C. et al. A transcriptomic method to determine airway immune dysfunction in T2-high and T2-low asthma. Am. J. Respir. Crit. Care Med. 199, 465–477 (2019).

    Article  CAS  Google Scholar 

  29. Singh, D. et al. COPD patients with chronic bronchitis and higher sputum eosinophil counts show increased type-2 and PDE4 gene expression in sputum. J. Cell. Mol. Med. 25, 905–918 (2021).

    Article  CAS  Google Scholar 

  30. Affar, E. B. et al. Essential dosage-dependent functions of the transcription factor yin yang 1 in late embryonic development and cell cycle progression. Mol. Cell. Biol. 26, 3565–3581 (2006).

    Article  Google Scholar 

  31. Takeuchi, J. K. et al. Chromatin remodelling complex dosage modulates transcription factor function in heart development. Nat. Commun. 2, 187 (2011).

    Article  Google Scholar 

  32. Sokolik, C. et al. Transcription factor competition allows embryonic stem cells to distinguish authentic signals from noise. Cell Syst. 1, 117–129 (2015).

    Article  CAS  Google Scholar 

  33. Bujalowski, W. & Lohman, T. M. Escherichia coli single-strand binding protein forms multiple, distinct complexes with single-stranded DNA. Biochemistry 25, 7799–7802 (1986).

    Article  CAS  Google Scholar 

  34. Lohman, T. M. & Overman, L. B. Two binding modes in Escherichia coli single strand binding protein-single stranded DNA complexes. Modulation by NaCl concentration. J. Biol. Chem. 260, 3594–3603 (1985).

    Article  CAS  Google Scholar 

  35. Reddy, M. S., Guhan, N. & Muniyappa, K. Characterization of single-stranded DNA-binding proteins from Mycobacteria. The carboxyl-terminal of domain of SSB is essential for stable association with its cognate RecA protein. J. Biol. Chem. 276, 45959–45968 (2001).

    Article  CAS  Google Scholar 

  36. Parks, B. GreenleafLab, matcha (GitHub, 2022); https://github.com/GreenleafLab/matcha

  37. Stuart, T. et al. Comprehensive integration of single-cell data. Cell 177, 1888–1902.e21 (2019).

    Article  CAS  Google Scholar 

  38. Granja, J. M. et al. Single-cell multiomic analysis identifies regulatory programs in mixed-phenotype acute leukemia. Nat. Biotechnol. 37, 1458–1465 (2019).

    Article  CAS  Google Scholar 

  39. Schep, A. N., Wu, B., Buenrostro, J. D. & Greenleaf, W. J. chromVAR: inferring transcription-factor-associated accessibility from single-cell epigenomic data. Nat. Methods 14, 975–978 (2017).

    Article  CAS  Google Scholar 

  40. Weirauch, M. T. et al. Determination and inference of eukaryotic transcription factor sequence specificity. Cell 158, 1431–1443 (2014).

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

  42. Li, H. et al. The Sequence Alignment/Map format and SAMtools. Bioinformatics 25, 2078–2079 (2009).

    Article  Google Scholar 

Download references

Acknowledgements

We thank R. Baskar, S. Pierce, S. Klemm, Y. Kim and members of the Greenleaf laboratory for helpful discussions and suggestions. We also thank the Stanford Fluorescence Activated Cell Sorting facility and Stanford Functional Genomics Facility for technical support. Figure schematics were created with BioRender.com. This work was supported by funding from the National Institutes of Health (NIH) (grant nos. P50HG007735, R01HG008140, R01HG009909, UM1HG011972, U01MH116529, U54HG010426, U01DK127419, U01HG011762, U19AI057266 and UM1HG009442), the Rita Allen Foundation, the Baxter Foundation Faculty Scholar Grant and the Human Frontiers Science Program (grant no. RGY006S) to W.J.G. W.J.G. is a Chan Zuckerberg Biohub investigator (grant nos. 2017-174468 and 2018-182817). Fellowship support was provided by the Stanford School of Medicine Dean’s Fellowship and NIH (grant no. F32GM135996) to A.F.C. and a training grant from the National Institute of Standards and Technology to B.P.

Author information

Authors and Affiliations

Authors

Contributions

A.F.C. and W.J.G. conceived the project and designed the experiments with input from all authors. A.F.C. led method development and performed experiments with help from A.S.K. A.F.C. and B.P. performed bioinformatic analysis, visualization and interpretation. J.J.G. provided input on the CD4 memory T cell experiment and interpretation of results. A.F.C. and W.J.G. drafted the manuscript. A.F.C., B.P., B.O.-R. and W.J.G. revised and edited the manuscript with input from all authors.

Corresponding author

Correspondence to William J. Greenleaf.

Ethics declarations

Competing interests

A.F.C. and W.J.G. are listed as coinventors on a patent related to this work. 10X Genomics holds the license to other patents in which W.J.G. is listed as an inventor. W.J.G. is an equity holder of 10X genomics and a co-founder of Protillion Biosciences. W.J.G. consults for Guardant Health, Quantapore, Protillion Biosciences, and Ultima Genomics. The remaining authors declare no competing interests.

Peer review

Peer review information

Nature Methods thanks the anonymous reviewers for their contribution to the peer review of this work. Lei Tang was the primary editor on this article and managed its editorial process and peer review in collaboration with the rest of the editorial 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 Staining of nuclear and cytosolic proteins using oligo-antibodies blocked with EcoSSB.

a) Sorting of cells expressing low, mid, or high levels of nuclear GFP that have been stained with an anti-GFP oligo-conjugated antibody. b) Quantitative PCR for the conjugated oligo from equal cell numbers of sorted populations in (a) for n = 2 technical replicates. c) Staining of K562 cells and mouse ESCs for endogenous GATA1 protein using an anti-GATA1 antibody linked to an 80 bp oligo with 3’-Cy5. d) Flow cytometry plot of HEK293T cells expressing cytosolic GFP and stained with an anti-GFP antibody linked to a 100 bp single stranded DNA oligo with 3’-Cy5 modification. e) Flow cytometry plot of nuclear GFP-expressing HEK293T cells with a GFP antibody linked to a Cy5-modified ssDNA oligo using the conditions indicated. “NEAT-seq”: NEAT-seq fixation, permeabilization, and staining conditions using oligo-antibodies pre-incubated with EcoSSB. “Dextran sulfate block”: NEAT-seq fixation and permeabilization conditions with inCITE-seq staining conditions (i.e with dextran sulfate blocking agent). “inCITE-seq”: inCITE-seq fixation, permeabilization, and staining conditions. Spearman correlation is shown.

Extended Data Fig. 2 NEAT-seq species mixing experiment data quality.

a-b) Cutoffs for annotating a cell as human, mouse, or mixed based on percentage of reads in a cell mapping to the human genome for ATAC-seq (a) and RNA-seq (b) data. c) Scatterplot of number of ATAC-seq reads mapping to the human vs mouse genome in each cell prior to removing HTO doublets, with each cell colored by its classification as a human cell, mouse cell, mixed species doublet, or an HTO doublet. d) Same as (c) but for RNA-seq reads. e) Fragment length distribution of ATAC-seq data generated using NEAT-seq. f) Average Tn5 insertions across transcriptional start sites normalized to the flanking region + /− 2 kb from the start site (i.e TSS enrichment) from scATAC-seq data generated using NEAT-seq. g) Log-transformed Tn5 insertions in ATAC-seq peaks for NEAT-seq data vs bulk ATAC-seq data in K562 cells. Pearson correlation is shown. h) A comparison of RNA-seq counts from bulk data vs NEAT-seq in K562 cells (left) or standard 10X Multiome data in GM12878 cells (right). Values are log-transformed TPM (for single cell data) or FPKM (for bulk data). Pearson correlation is shown. i) Western blot of mESC and K562 cell lysate using oligo-conjugated SOX2 antibody pre-incubated with EcoSSB and detected with IR800 secondary antibody (Licor). GAPDH was also probed as a loading control with IR700 secondary antibody. Imaging was performed on a Licor Odyssey imaging system. Images shown are representative results of 2 independent experiments.

Source data

Extended Data Fig. 3 CD4 memory T cell subset annotations.

a) scRNA-seq UMAP of CD4 memory T cells colored with scATAC-seq cluster IDs. TCM = central memory, Act. = recently activated cells, Uncom. = uncommitted memory cells. b) RNA expression of master TF drivers and canonical cell surface markers of CD4 memory cell subsets in each scATAC-seq cluster. TBX21 = Tbet transcript, RORC = RORγT transcript, IKZF2 = Helios transcript. c) Gene accessibility for cytokines induced in different CD4 T cell subsets overlayed on the scATAC-seq UMAP. d) RNA levels for the cytokines in (c). e) chromVAR deviation scores for the naïve and CM T cell TFs, LEF1 and TCF7, overlayed on scATAC-seq UMAP. f) RNA expression of the CM marker, CCR7, overlayed on the scATAC-seq UMAP.

Extended Data Fig. 4 Enrichment of TF ADTs in the T cell subtype driven by the TF.

a) Unsmoothed ADT counts for each TF overlayed on the scATAC-seq UMAP after the indicated normalization method (raw counts, NPC normalization, and total ADT counts normalization). b) Log2-transformed, NPC-normalized ADT counts for each TF separated by scATAC-seq cluster for cells stained with antibody concentration 1 (see methods). c) Same as (b) but for antibody concentration 2. d) Scatterplots with marginal histograms of log2-transformed read-normalized RNA vs log2-transformed NPC-normalized ADT counts for each TF. Colored data points represent cells belonging to the scATAC-seq cluster most enriched in expression of the indicated TF. e) Unsmoothed, normalized RNA counts of the indicated TFs overlayed on the scATAC-seq UMAP.

Extended Data Fig. 5 Correlation of ADT levels with gene locus accessibility, RNA, and motif accessibility for each TF.

a) Correlation across all cells for each measurement. Values were first smoothed across neighboring cells using MAGIC imputation to account for dropouts. Pearson correlations are shown. b) The data in (a) but averaged across cells within each scATAC-seq cluster.

Extended Data Fig. 6 Post-transcriptional regulation of GATA3.

a) Flow cytometry plots of GATA3-transfected and WT HEK293T cells stained with an EcoSSB-bound oligo-conjugated GATA3 antibody, using a fluorescent secondary antibody for detection. b) Log2-transformed RNA levels of the indicated gene in cells expressing high RNA and low protein (“low”) vs high RNA and high protein for GATA3 (“high”). The mean RNA expression for each group is shown above the violin plot. N = 140 cells examined over one independent experiment for both “high” and “low” populations. Boxplots show median with bounds of the box representing the 25th and 75th percentiles and the whiskers extending to the value closest to but not exceeding 1.5 times the interquartile range. Data extending beyond the whiskers are plotted individually as outliers.

Extended Data Fig. 7 Identification of DNA regulatory elements and genes correlated with master TF protein expression.

a) Spearman correlations between ATAC-seq peak accessibility and NPC-normalized TF ADT counts across single cells. Cutoffs for significant correlations are indicated by dashed lines (see Methods). Points in red indicate peaks containing a binding motif for the TF. TF motif enrichment in significantly correlated peaks was calculated using a hypergeometric test. b) Spearman correlations between read-normalized RNA counts and NPC-normalized TF ADT counts across single cells. Cutoffs for significant correlations are indicated by dashed lines (see Methods). Significantly correlated genes known to be enriched or play a functional role in the relevant T cell subset are labeled. c) CCR4 ATAC-seq tracks in CD4 memory cells separated into quintiles by GATA3 ADT levels, along with significantly correlated peak-gene linkages (adj. p < 0.05). Spearman correlations are shown. Peaks containing a GATA3 motif are indicated. d-e) CCR4 RNA expression (d) and accessibility at the highlighted GATA3 motif-containing peak (e) as a function of GATA3 ADT levels. Mean is shown with standard error of the mean of n = 768 + /− 1 cells per group.

Extended Data Fig. 8 GO term enrichment for candidate target genes in TF-driven peak-gene linkages.

Enriched GO terms in the target gene list were identified by hypergeometric test using enrichGO in the clusterProfiler R package, using all genes with at least one RNA count in the dataset as a background gene list. Adjusted p-values were calculated using the Benjamini-Hochberg procedure.

Extended Data Fig. 9 Single-tissue eQTL analysis for the rs62088464-TSEN54 variant-gene pair from GTEx portal.

a) Normalized effect size (NES) across various tissues for the protective (G) vs risk (A) allele on TSEN54 expression. The risk allele preserves the GATA3 motif. A negative NES value indicates a gene with expression that is associated with the risk allele. Error bars indicate 95% confidence intervals. b) Normalized TSEN54 expression grouped by rs62088464 genotype for lung and esophagus mucosa. c) Genes ordered by NES across tissues for rs62088464. P-values for (b) and (c) are outputs from the GTEx portal (release v8) and are calculated from a two-sided t-test comparing the observed NES in a tissue to a null NES of 0.

Supplementary information

Supplementary Information

Supplementary Notes 1–3 and Protocol.

Reporting Summary

Supplementary Table

Supplementary Tables 1–4.

Source data

Source Data Extended Data Fig. 2

Unprocessed western blot for panel Fig. 2i.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Chen, A.F., Parks, B., Kathiria, A.S. et al. NEAT-seq: simultaneous profiling of intra-nuclear proteins, chromatin accessibility and gene expression in single cells. Nat Methods 19, 547–553 (2022). https://doi.org/10.1038/s41592-022-01461-y

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41592-022-01461-y

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