nature.com

Spatially resolved genome-wide joint profiling of epigenome and transcriptome with spatial-Atac-RNA-seq and…

Abstract

The epigenome of a cell is tightly correlated with gene transcription, which controls cell identity and diverse biological activities. Recent advances in spatial technologies have improved our understanding of tissue heterogeneity by analyzing transcriptomics or epigenomics with spatial information preserved, but have been mainly restricted to one molecular layer at a time. Here we present procedures for two spatially resolved sequencing methods, spatial-ATAC-RNA-seq and spatial-CUT&Tag-RNA-seq, that co-profile transcriptome and epigenome genome wide. In both methods, transcriptomic readouts are generated through tissue fixation, permeabilization and in situ reverse transcription. In spatial-ATAC-RNA-seq, Tn5 transposase is used to probe accessible chromatin, and in spatial-CUT&Tag-RNA-seq, the tissue is incubated with primary antibodies that target histone modifications, followed by Protein A-fused Tn5-induced tagmentation. Both methods leverage a microfluidic device that delivers two sets of oligonucleotide barcodes to generate a two-dimensional mosaic of tissue pixels at near single-cell resolution. A spatial-ATAC-RNA-seq or spatial-CUT&Tag-RNA-seq library can be generated in 3–5 d, allowing researchers to simultaneously investigate the transcriptomic landscape and epigenomic landscape of an intact tissue section. This protocol is an extension of our previous spatially resolved epigenome sequencing protocol and provides opportunities in multimodal profiling.

Key points

Spatially resolved concurrent profiling of both transcriptome and epigenome is essential for studying spatio-temporal regulation of gene expression, but the technical solutions permitting such analyses remain limited.

This protocol describes spatial-ATAC-RNA-seq and spatial-CUT&Tag-RNA-seq, which profile genome-wide transcription jointly with open chromatin and histone modifications, respectively, on a tissue section.

This is a preview of subscription content, access via your institution

Access options

Access through your institution

Change institution

Buy or subscribe

Access Nature and 54 other Nature Portfolio journals

Get Nature+, our best-value online-access subscription

$29.99 / 30 days

cancel any time

Learn more

Subscribe to this journal

Receive 12 print issues and online access

$259.00 per year

only $21.58 per issue

Learn more

Buy this article

Purchase on SpringerLink

Instant access to full article PDF

Buy now

Prices may be subject to local taxes which are calculated during checkout

Additional access options:

Log in

Learn about institutional subscriptions

Read our FAQs

Contact customer support

Fig. 1: Overview of the procedures for spatial-ATAC-RNA-seq and spatial-CUT&Tag-RNA-seq.

Fig. 2: Library structure and oligonucleotide primer sequence design.

Fig. 3: Procedures for tissue fixation, permeabilization and Tn5 or pA-Tn5 tagmentation.

Fig. 4: Workflow of deterministic barcoding in tissue.

Fig. 5: Anticipated results of library visualization.

Fig. 6: Bioinformatics pipelines for analyzing spatially resolved epigenome-transcriptome data.

Fig. 7: Spatial-ATAC-RNA-seq and spatial-CUT&Tag-RNA-seq data quality control.

Fig. 8: Evaluation and analysis of a spatial-ATAC-RNA-seq dataset.

Data availability

Original data that are used to generate metrics plots in Figs. 7 and 8 are available in Source Data. Raw data for the illustrative results shown in this protocol are available in Zhang et al.26. Source data are provided with this paper.

Code availability

Codes for processing data of the associated publication26 are available at https://github.com/di-0579/Spatial_epigenome-transcriptome_co-sequencing.

References

Klemm, S. L., Shipony, Z. & Greenleaf, W. J. Chromatin accessibility and the regulatory epigenome. Nat. Rev. Genet. 20, 207–220 (2019).

CASPubMedGoogle Scholar

Bannister, A. J. & Kouzarides, T. Regulation of chromatin by histone modifications. Cell Res. 21, 381–395 (2011).

CASPubMedPubMed CentralGoogle Scholar

Fleck, J. S. et al. Inferring and perturbing cell fate regulomes in human brain organoids. Nature 621, 365–372 (2022).

PubMedPubMed CentralGoogle Scholar

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

CASPubMedPubMed CentralGoogle Scholar

Li, H. et al. Transcriptomic, epigenomic, and spatial metabolomic cell profiling redefines regional human kidney anatomy. Cell Metab. 36, 1105–1125.e10 (2024).

CASPubMedGoogle Scholar

Wang, G. et al. Integrating genetics with single-cell multiomic measurements across disease states identifies mechanisms of beta cell dysfunction in type 2 diabetes. Nat. Genet. 55, 984–994 (2023).

CASPubMedPubMed CentralGoogle Scholar

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

CASPubMedPubMed CentralGoogle Scholar

Baysoy, A., Bai, Z., Satija, R. & Fan, R. The technological landscape and applications of single-cell multi-omics. Nat. Rev. Mol. Cell Biol. 24, 695–713 (2023).

CASPubMedGoogle Scholar

Muto, Y., Li, H. & Humphreys, B. D. in Innovations in Nephrology: Breakthrough Technologies in Kidney Disease Care 87–102 (Springer, 2022); https://doi.org/10.1007/978-3-031-11570-7_5

Rao, A., Barkley, D., França, G. S. & Yanai, I. Exploring tissue architecture using spatial transcriptomics. Nature 596, 211–220 (2021).

CASPubMedPubMed CentralGoogle Scholar

Rodriques, S. G. et al. Slide-seq: a scalable technology for measuring genome-wide expression at high spatial resolution. Science 363, 1463–1467 (2019).

CASPubMedPubMed CentralGoogle Scholar

Lu, T., Ang, C. E. & Zhuang, X. Spatially resolved epigenomic profiling of single cells in complex tissues. Cell 185, 4448–4464.e17 (2022).

CASPubMedPubMed CentralGoogle Scholar

Chen, A. et al. Spatiotemporal transcriptomic atlas of mouse organogenesis using DNA nanoball-patterned arrays. Cell 185, 1777–1792.e21 (2022).

CASPubMedGoogle Scholar

Liu, Y. et al. High-plex protein and whole transcriptome co-mapping at cellular resolution with spatial CITE-seq. Nat. Biotechnol. https://doi.org/10.1038/s41587-023-01676-0 (2023).

Liu, Y. et al. High-spatial-resolution multi-omics sequencing via deterministic barcoding in tissue. Cell 183, 1665–1681.e18 (2020).

CASPubMedPubMed CentralGoogle Scholar

Wirth, J. et al. Spatial transcriptomics using multiplexed deterministic barcoding in tissue. Nat. Commun. 14, 1–15 (2023).

CASGoogle Scholar

Jiang, F. et al. Simultaneous profiling of spatial gene expression and chromatin accessibility during mouse brain development. Nat. Methods 20, 1048–1057 (2023).

CASPubMedGoogle Scholar

Buenrostro, J. D., Giresi, P. G., Zaba, L. C., Chang, H. Y. & Greenleaf, W. J. Transposition of native chromatin for fast and sensitive epigenomic profiling of open chromatin, DNA-binding proteins and nucleosome position. Nat. Methods 10, 1213–1218 (2013).

CASPubMedPubMed CentralGoogle Scholar

Xu, W. et al. A plate-based single-cell ATAC-seq workflow for fast and robust profiling of chromatin accessibility. Nat. Protoc. 16, 4084–4107 (2021).

CASPubMedGoogle Scholar

Grandi, F. C., Modi, H., Kampman, L. & Corces, M. R. Chromatin accessibility profiling by ATAC-seq. Nat. Protoc. 17, 1518–1552 (2022).

CASPubMedPubMed CentralGoogle Scholar

Kaya-Okur, H. S. et al. CUT&Tag for efficient epigenomic profiling of small samples and single cells. Nat. Commun. 10, 1930 (2019).

PubMedPubMed CentralGoogle Scholar

Kaya-Okur, H. S., Janssens, D. H., Henikoff, J. G., Ahmad, K. & Henikoff, S. Efficient low-cost chromatin profiling with CUT&Tag. Nat. Protoc. 15, 3264–3283 (2020).

CASPubMedPubMed CentralGoogle Scholar

Deng, Y. et al. Spatial-CUT&Tag: spatially resolved chromatin modification profiling at the cellular level. Science 375, 681–686 (2022).

CASPubMedPubMed CentralGoogle Scholar

Deng, Y. et al. Spatial profiling of chromatin accessibility in mouse and human tissues. Nature 609, 375–383 (2022).

CASPubMedPubMed CentralGoogle Scholar

Farzad, N. et al. Spatially resolved epigenome sequencing via Tn5 transposition and deterministic DNA barcoding in tissue. Nat. Protoc. 19, 3389–3425 (2024).

CASPubMedGoogle Scholar

Zhang, D. et al. Spatial epigenome–transcriptome co-profiling of mammalian tissues. Nature 616, 113–122 (2023).

CASPubMedPubMed CentralGoogle Scholar

Wimmers, F. et al. Multi-omics analysis of mucosal and systemic immunity to SARS-CoV-2 after birth. Cell 186, 4632–4651.e23 (2023).

CASPubMedPubMed CentralGoogle Scholar

Terekhanova, N. V. et al. Epigenetic regulation during cancer transitions across 11 tumour types. Nature 623, 432–441 (2023).

CASPubMedPubMed CentralGoogle Scholar

Suppinger, S. et al. Multimodal characterization of murine gastruloid development. Cell Stem Cell 30, 867–884.e11 (2023).

CASPubMedPubMed CentralGoogle Scholar

Li, H. & Humphreys, B. D. Multimodal characterization of sexual dimorphism in the mammalian kidney. Kidney Int. 105, 653–655 (2024).

CASPubMedGoogle Scholar

Bartosovic, M., Kabbe, M. & Castelo-Branco, G. Single-cell CUT&Tag profiles histone modifications and transcription factors in complex tissues. Nat. Biotechnol. 39, 825–835 (2021).

CASPubMedPubMed CentralGoogle Scholar

Fu, Z. et al. Cut&tag: a powerful epigenetic tool for chromatin profiling. Epigenetics 19, 2293411 (2024).

PubMedGoogle Scholar

Wu, S. J. et al. Single-cell CUT&Tag analysis of chromatin modifications in differentiation and tumor progression. Nat. Biotechnol. 39, 819–824 (2021).

CASPubMedPubMed CentralGoogle Scholar

Janssens, D. H. et al. Scalable single-cell profiling of chromatin modifications with sciCUT&Tag. Nat. Protoc. 19, 83–112 (2023).

PubMedPubMed CentralGoogle Scholar

Li, H., Li, D. & Humphreys, B. D. Chromatin conformation and histone modification profiling across human kidney anatomic regions. Sci. Data 11, 1–12 (2024).

Google Scholar

Kamimoto, K. et al. Dissecting cell identity via network inference and in silico gene perturbation. Nature 614, 742–751 (2023).

CASPubMedPubMed CentralGoogle Scholar

Zhang, D. et al. Spatial dynamics of mammalian brain development and neuroinflammation by multimodal tri-omics mapping. Preprint at bioRxivhttps://doi.org/10.1101/2024.07.28.605493 (2024).

Lee, M. Y. Y., Kaestner, K. H. & Li, M. Benchmarking algorithms for joint integration of unpaired and paired single-cell RNA-seq and ATAC-seq data. Genome Biol. 24, 244 (2023).

PubMedPubMed CentralGoogle Scholar

Ledru, N. et al. Predicting proximal tubule failed repair drivers through regularized regression analysis of single cell multiomic sequencing. Nat. Commun. 15, 1–19 (2024).

Google Scholar

Stuart, T. & Satija, R. Integrative single-cell analysis. Nat. Rev. Genet. 20, 257–272 (2019).

CASPubMedGoogle Scholar

Luecken, M. D. et al. Benchmarking atlas-level data integration in single-cell genomics. Nat. Methods 19, 41–50 (2021).

PubMedPubMed CentralGoogle Scholar

Stickels, R. R. et al. Highly sensitive spatial transcriptomics at near-cellular resolution with Slide-seqV2. Nat. Biotechnol. 39, 313–319 (2020).

PubMedPubMed CentralGoogle Scholar

Vickovic, S. et al. High-definition spatial transcriptomics for in situ tissue profiling. Nat. Methods 16, 987 (2019).

CASPubMedPubMed CentralGoogle Scholar

Llorens-Bobadilla, E. et al. Solid-phase capture and profiling of open chromatin by spatial ATAC. Nat. Biotechnol. 41, 1085–1088 (2023).

CASPubMedPubMed CentralGoogle Scholar

Russell, A. J. C. et al. Slide-tags enables single-nucleus barcoding for multimodal spatial genomics. Nature 625, 101–109 (2023).

PubMedPubMed CentralGoogle Scholar

Li, H. et al. A comprehensive benchmarking with practical guidelines for cellular deconvolution of spatial transcriptomics. Nat. Commun. 14, 1548 (2023).

PubMedPubMed CentralGoogle Scholar

Bai, Z. et al. Spatially exploring RNA biology in archival formalin-fixed paraffin-embedded tissues. Cell 187, 6760–6779.e24 (2024).

CASPubMedGoogle Scholar

Kvastad, L. et al. The spatial RNA integrity number assay for in situ evaluation of transcriptome quality. Commun. Biol. 4, 57 (2021).

CASPubMedPubMed CentralGoogle Scholar

Adey, A. C. Tagmentation-based single-cell genomics. Genome Res. 31, 1693 (2021).

CASPubMedPubMed CentralGoogle Scholar

Picelli, S. et al. Tn5 transposase and tagmentation procedures for massively scaled sequencing projects. Genome Res. 24, 2033–2040 (2014).

CASPubMedPubMed CentralGoogle Scholar

Li, H. & Humphreys, B. D. Protocol for multimodal profiling of human kidneys with simultaneous high-throughput ATAC and RNA expression with sequencing. STAR Protoc. 5, 103049 (2024).

CASPubMedPubMed CentralGoogle Scholar

Ramsköld, D. et al. Full-length mRNA-seq from single-cell levels of RNA and individual circulating tumor cells. Nat. Biotechnol. 30, 777–782 (2012).

PubMedPubMed CentralGoogle Scholar

Wulf, M. G. et al. Non-templated addition and template switching by Moloney murine leukemia virus (MMLV)-based reverse transcriptases co-occur and compete with each other. J. Biol. Chem. 294, 18220 (2019).

CASPubMedPubMed CentralGoogle Scholar

Li, H. & Humphreys, B. D. Mouse kidney nuclear isolation and library preparation for single-cell combinatorial indexing RNA sequencing. STAR Protoc. 3, 101904 (2022).

CASPubMedPubMed CentralGoogle Scholar

Su, G. et al. Spatial multi-omics sequencing for fixed tissue via DBiT-seq. STAR Protoc. 2, 100532 (2021).

CASPubMedPubMed CentralGoogle Scholar

Satpathy, A. T. et al. Massively parallel single-cell chromatin landscapes of human immune cell development and intratumoral T cell exhaustion. Nat. Biotechnol. 37, 925–936 (2019).

CASPubMedPubMed CentralGoogle Scholar

Navarro, J. F., Sjöstrand, J., Salmén, F., Lundeberg, J. & Ståhl, P. L. ST Pipeline: an automated pipeline for spatial mapping of unique transcripts. Bioinformatics 33, 2591–2593 (2017).

PubMedGoogle Scholar

Stuart, T., Srivastava, A., Madad, S., Lareau, C. A. & Satija, R. Single-cell chromatin state analysis with Signac. Nat. Methods 18, 1333–1341 (2021).

CASPubMedPubMed CentralGoogle Scholar

Granja, J. M. et al. ArchR is a scalable software package for integrative single-cell chromatin accessibility analysis. Nat. Genet. 53, 403–411 (2021).

CASPubMedPubMed CentralGoogle Scholar

Zhang, K., Zemke, N. R., Armand, E. J. & Ren, B. A fast, scalable and versatile tool for analysis of single-cell omics data. Nat. Methods 21, 217–227 (2024).

CASPubMedPubMed CentralGoogle Scholar

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

CASPubMedPubMed CentralGoogle Scholar

Cao, Z. J. & Gao, G. Multi-omics single-cell data integration and regulatory inference with graph-linked embedding. Nat. Biotechnol. 40, 1458–1466 (2022).

CASPubMedPubMed CentralGoogle Scholar

Yao, Z. et al. A high-resolution transcriptomic and spatial atlas of cell types in the whole mouse brain. Nature 624, 317–332 (2023).

CASPubMedPubMed CentralGoogle Scholar

Wolf, F. A., Angerer, P. & Theis, F. J. SCANPY: large-scale single-cell gene expression data analysis. Genome Biol. 19, 15 (2018).

PubMedPubMed CentralGoogle Scholar

Download references

Acknowledgements

We thank the Yale Center for Research Computing for guidance and use of the research computing infrastructure. The molds for microfluidic devices were fabricated at the Yale University School of Engineering and Applied Science Nanofabrication Center. Next-generation sequencing was conducted at the Yale Center for Genome Analysis, as well as at the Yale Stem Cell Center Genomics Core Facility, which was supported by the Connecticut Regenerative Medicine Research Fund and the Li Ka Shing Foundation. A service provided by the Genomics Core of Yale Cooperative Center of Excellence in Hematology (U54DK106857) was used. This research was supported by Packard Fellowship for Science and Engineering (to R.F.), Yale Stem Cell Center Chen Innovation Award (to R.F.), and grants from the US National Institutes of Health (U54AG076043, U54AG079759, UG3CA257393, UH3CA257393, R01CA245313, RF1MH128876, U54CA274509, U54CA268083 and U01CA294514 to R.F.). Illustrations were created with BioRender.com.

Author information

Authors and Affiliations

Department of Biomedical Engineering, Yale University, New Haven, CT, USA

Haikuo Li, Shuozhen Bao, Negin Farzad, Xiaoyu Qin, Anthony A. Fung, Di Zhang, Zhiliang Bai & Rong Fan

Department of Pathology, Yale University School of Medicine, New Haven, CT, USA

Bo Tao & Rong Fan

Yale Stem Cell Center and Yale Cancer Center, Yale University School of Medicine, New Haven, CT, USA

Rong Fan

Yale Center for Research on Aging (Y-Age), Yale University School of Medicine, New Haven, CT, USA

Rong Fan

Authors

Haikuo Li

View author publications

You can also search for this author inPubMedGoogle Scholar

2. Shuozhen Bao

View author publications

You can also search for this author inPubMedGoogle Scholar

3. Negin Farzad

View author publications

You can also search for this author inPubMedGoogle Scholar

4. Xiaoyu Qin

View author publications

You can also search for this author inPubMedGoogle Scholar

5. Anthony A. Fung

View author publications

You can also search for this author inPubMedGoogle Scholar

6. Di Zhang

View author publications

You can also search for this author inPubMedGoogle Scholar

7. Zhiliang Bai

View author publications

You can also search for this author inPubMedGoogle Scholar

8. Bo Tao

View author publications

You can also search for this author inPubMedGoogle Scholar

9. Rong Fan

View author publications

You can also search for this author inPubMedGoogle Scholar

Contributions

H.L., N.F. and R.F. conceived, coordinated and designed the study. H.L., S.B., X.Q. and B.T. wrote the manuscript. H.L., S.B., A.A.F., D.Z., Z.B. and B.T. analyzed data and created figures. R.F. supervised the project and revised the manuscript. All authors read and approved the final manuscript.

Corresponding author

Correspondence to Rong Fan.

Ethics declarations

Competing interests

R.F. is scientific founder of and advisor to IsoPlexis, Singleron Biotechnologies and AtlasXomics. The Yale University Provost’s Office reviewed and managed the interests of R.F. in accordance with the University’s conflict of interest policies. The other authors declare no competing interests.

Peer review

Peer review information

Nature Reviews thanks the anonymous reviewer(s) for their contribution to the peer review of this work.

Additional information

Key references

Zhang, D. et al. Spatial epigenome–transcriptome co-profiling of mammalian tissues. Nature 616, 113–122 (2023): https://doi.org/10.1038/s41586-023-05795-1

Farzad, N. et al. Spatially resolved epigenome sequencing via Tn5 transposition and deterministic DNA barcoding in tissue. Nat. Protoc. 19, 3389–3425 (2024): https://doi.org/10.1038/s41596-024-01013-y

This protocol is an extension to: Nat. Protoc. 19, 3389–3425 (2024): https://doi.org/10.1038/s41596-024-01013-y

Extended data

Extended Data Fig. 1 Anticipated results of spatial-ATAC-seq library visualization.

TapeStation D5000 electropherogram (High-Sensitivity) showing fragment distribution of a spatial-ATAC-seq library generated from a human brain sample. The table on the right panel indicates the concentration, molarity and fractions of fragments in the select region (200-5,000 bp).

Supplementary information

Reporting Summary

Supplementary Table 1

Oligo sequence information.

Supplementary Data 1

Microfluidic CAD designs.

Supplementary Video 1

Video illustration for attaching PDMS reservoirs on the tissue.

Supplementary Video 2

Video illustration for ATAC-seq and CUT&Tag related procedures.

Supplementary Video 3

Video illustration for attaching PDMS chips on the tissue.

Supplementary Video 4

Video illustration for streptavidin-bead affinity pulldown.

Supplementary Video 5

Video illustration for PDMS associated equipment preparation.

Source data

Source Data Figs. 7 and 8

Original data used to generate metrics plots in Figs. 7 and 8.

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

Li, H., Bao, S., Farzad, N. et al. Spatially resolved genome-wide joint profiling of epigenome and transcriptome with spatial-ATAC-RNA-seq and spatial-CUT&Tag-RNA-seq. Nat Protoc (2025). https://doi.org/10.1038/s41596-025-01145-9

Download citation

Received:29 June 2024

Accepted:15 November 2024

Published:21 March 2025

DOI:https://doi.org/10.1038/s41596-025-01145-9

Share this article

Anyone you share the following link with will be able to read this content:

Get shareable link

Sorry, a shareable link is not currently available for this article.

Copy to clipboard

Provided by the Springer Nature SharedIt content-sharing initiative

Read full news in source page