Abstract
T cells create vast amounts of diversity in the genes that encode their T cell receptors (TCRs), which enables individual clones to recognize specific peptide–major histocompatibility complex (MHC) ligands. Here we combined sequencing of the TCR-encoding genes with assay for transposase-accessible chromatin with sequencing (ATAC-seq) analysis at the single-cell level to provide information on the TCR specificity and epigenomic state of individual T cells. By using this approach, termed transcript-indexed ATAC-seq (T-ATAC-seq), we identified epigenomic signatures in immortalized leukemic T cells, primary human T cells from healthy volunteers and primary leukemic T cells from patient samples. In peripheral blood CD4+ T cells from healthy individuals, we identified cis and trans regulators of naive and memory T cell states and found substantial heterogeneity in surface-marker-defined T cell populations. In patients with a leukemic form of cutaneous T cell lymphoma, T-ATAC-seq enabled identification of leukemic and nonleukemic regulatory pathways in T cells from the same individual by allowing separation of the signals that arose from the malignant clone from the background T cell noise. Thus, T-ATAC-seq is a new tool that enables analysis of epigenomic landscapes in clonal T cells and should be valuable for studies of T cell malignancy, immunity and immunotherapy.
This is a preview of subscription content, access via your institution
Relevant articles
Open Access articles citing this article.
-
Single-cell technologies in multiple myeloma: new insights into disease pathogenesis and translational implications
Biomarker Research Open Access 31 May 2023
-
Epigenetic remodeling of the immune landscape in cancer: therapeutic hurdles and opportunities
Journal of Biomedical Science Open Access 10 January 2023
-
A roadmap for translational cancer glycoimmunology at single cell resolution
Journal of Experimental & Clinical Cancer Research Open Access 15 April 2022
Access options
Access Nature and 54 other Nature Portfolio journals
Get Nature+, our best-value online-access subscription
$29.99 / 30 days
cancel any time
Subscribe to this journal
Receive 12 print issues and online access
$209.00 per year
only $17.42 per issue
Rent or buy this article
Prices vary by article type
from$1.95
to$39.95
Prices may be subject to local taxes which are calculated during checkout





References
Davis, M. M. & Bjorkman, P. J. T cell antigen receptor genes and T cell recognition. Nature 334, 395–402 (1988).
Shalek, A. K. et al. Single-cell RNA-seq reveals dynamic paracrine control of cellular variation. Nature 510, 363–369 (2014).
Gaublomme, J. T. et al. Single-cell genomics unveils critical regulators of TH17 cell pathogenicity. Cell 163, 1400–1412 (2015).
Paul, F. et al. Transcriptional heterogeneity and lineage commitment in myeloid progenitors. Cell 163, 1663–1677 (2015).
Tirosh, I. et al. Dissecting the multicellular ecosystem of metastatic melanoma by single-cell RNA-seq. Science 352, 189–196 (2016).
Han, A., Glanville, J., Hansmann, L. & Davis, M. M. Linking T cell receptor sequence to functional phenotype at the single-cell level. Nat. Biotechnol. 32, 684–692 (2014).
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).
Buenrostro, J. D. et al. Single-cell chromatin accessibility reveals principles of regulatory variation. Nature 523, 486–490 (2015).
Corces, M. R. et al. Lineage-specific and single-cell chromatin accessibility charts human hematopoiesis and leukemia evolution. Nat. Genet. 48, 1193–1203 (2016).
Buenrostro, J. D. et al. Single-cell epigenomics maps the continuous regulatory landscape of human hematopoietic differentiation. bioRxiv http://dx.doi.org/10.1101/109843 (2017).
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).
Stubbington, M. J. T. et al. T cell fate and clonality inference from single-cell transcriptomes. Nat. Methods 13, 329–332 (2016).
Afik, S. et al. Targeted reconstruction of T cell receptor sequence from single-cell RNA-seq links CDR3 length to T cell differentiation state. Nucleic Acids Res. 45, e148 (2017).
Cusanovich, D. A. et al. Multiplex single-cell profiling of chromatin accessibility by combinatorial cellular indexing. Science 348, 910–914 (2015).
Weber, B. N. et al. A critical role for TCF-1 in T lineage specification and differentiation. Nature 476, 63–68 (2011).
Collins, A., Littman, D. R. & Taniuchi, I. RUNX proteins in transcription factor networks that regulate T cell lineage choice. Nat. Rev. Immunol. 9, 106–115 (2009).
Morita, R. et al. Human blood CXCR5+CD4+ T cells are counterparts of T follicular cells and contain specific subsets that differentially support antibody secretion. Immunity 34, 108–121 (2011).
Fontenot, J. D., Rasmussen, J. P., Gavin, M. A. & Rudensky, A. Y. A function for interleukin 2 in Foxp3-expressing regulatory T cells. Nat. Immunol. 6, 1142–1151 (2005).
Ouyang, W., Kolls, J. K. & Zheng, Y. The biological functions of T helper 17 cell effector cytokines in inflammation. Immunity 28, 454–467 (2008).
Meller, S. et al. TH17 cells promote microbial killing and innate immune sensing of DNA via interleukin 26. Nat. Immunol. 16, 970–979 (2015).
van der Maaten, L. & Hinton, G. Visualizing data using t-SNE. J. Mach. Learn. Res. 9, 2579–2605 (2008).
Kimmig, S. et al. Two subsets of naive T helper cells with distinct T cell receptor excision circle content in human adult peripheral blood. J. Exp. Med. 195, 789–794 (2002).
Boursalian, T. E., Golob, J., Soper, D. M., Cooper, C. J. & Fink, P. J. Continued maturation of thymic emigrants in the periphery. Nat. Immunol. 5, 418–425 (2004).
Harari, A., Vallelian, F. & Pantaleo, G. Phenotypic heterogeneity of antigen-specific CD4 T cells under different conditions of antigen persistence and antigen load. Eur. J. Immunol. 34, 3525–3533 (2004).
Zhao, C. & Davies, J. D. A peripheral CD4+ T cell precursor for naive, memory and regulatory T cells. J. Exp. Med. 207, 2883–2894 (2010).
Song, K. et al. Characterization of subsets of CD4+ memory T cells reveals early branched pathways of T cell differentiation in humans. Proc. Natl Acad. Sci. USA 102, 7916–7921 (2005).
Gattinoni, L. et al. A human memory T cell subset with stem-cell-like properties. Nat. Med. 17, 1290–1297 (2011).
Weiskopf, D. et al. Dengue virus infection elicits highly polarized CX3CR1+ cytotoxic CD4+ T cells associated with protective immunity. Proc. Natl Acad. Sci. USA 112, E4256–E4263 (2015).
Yui, M. A. & Rothenberg, E. V. Developmental gene networks: a triathlon on the course to T cell identity. Nat. Rev. Immunol. 14, 529–545 (2014).
Zheng, W. & Flavell, R. A. The transcription factor GATA-3 is necessary and sufficient for TH2 cytokine gene expression in CD4 T cells. Cell 89, 587–596 (1997).
Lohoff, M. & Mak, T. W. Roles of interferon-regulatory factors in T helper cell differentiation. Nat. Rev. Immunol. 5, 125–135 (2005).
Ivanov, I. I. et al. The orphan nuclear receptor ROR-γt directs the differentiation program of proinflammatory IL-17+ T helper cells. Cell 126, 1121–1133 (2006).
Yang, X. O. et al. T helper 17 lineage differentiation is programmed by orphan nuclear receptors ROR-α and ROR-γ. Immunity 28, 29–39 (2008).
Bauquet, A. T. et al. The co-stimulatory molecule ICOS regulates the expression of c-Maf and IL-21 in the development of follicular T helper cells and TH17 cells. Nat. Immunol. 10, 167–175 (2009).
Schraml, B. U. et al. The AP-1 transcription factor Batf controls TH17 differentiation. Nature 460, 405–409 (2009).
O’Shea, J. J., Lahesmaa, R., Vahedi, G., Laurence, A. & Kanno, Y. Genomic views of STAT function in CD4+ T helper cell differentiation. Nat. Rev. Immunol. 11, 239–250 (2011).
Rutz, S. et al. Transcription factor c-Maf mediates the TGF-β-dependent suppression of IL-22 production in TH17 cells. Nat. Immunol. 12, 1238–1245 (2011).
Ciofani, M. et al. A validated regulatory network for TH17 cell specification. Cell 151, 289–303 (2012).
Bigler, R. D., Boselli, C. M., Foley, B. & Vonderheid, E. C. Failure of anti–T cell receptor Vβ antibodies to consistently identify a malignant T cell clone in Sézary syndrome. Am. J. Pathol. 149, 1477–1483 (1996).
Kelemen, K., Guitart, J., Kuzel, T. M., Goolsby, C. L. & Peterson, L. C. The usefulness of CD26 in flow cytometric analysis of peripheral blood in Sézary syndrome. Am. J. Clin. Pathol. 129, 146–156 (2008).
Weng, W. K. et al. Minimal residual disease monitoring with high-throughput sequencing of T cell receptors in cutaneous T cell lymphoma. Sci. Transl. Med. 5, 214ra171 (2013).
Sufficool, K. E. et al. T cell clonality assessment by next-generation sequencing improves detection sensitivity in mycosis fungoides. J. Am. Acad. Dermatol. 73, 228–236 (2015).
Rook, A. H., Vowels, B. R., Jaworsky, C., Singh, A. & Lessin, S. R. The immunopathogenesis of cutaneous T cell lymphoma. Abnormal cytokine production by Sézary T cells. Arch. Dermatol. 129, 486–489 (1993).
Vowels, B. R. et al. TH2 cytokine mRNA expression in skin in cutaneous T cell lymphoma. J. Invest. Dermatol. 103, 669–673 (1994).
Krejsgaard, T., Odum, N., Geisler, C., Wasik, M. A. & Woetmann, A. Regulatory T cells and immunodeficiency in mycosis fungoides and Sézary syndrome. Leukemia 26, 424–432 (2012).
Ungewickell, A. et al. Genomic analysis of mycosis fungoides and Sézary syndrome identifies recurrent alterations in TNFR2. Nat. Genet. 47, 1056–1060 (2015).
Choi, J. et al. Genomic landscape of cutaneous T cell lymphoma. Nat. Genet. 47, 1011–1019 (2015).
Bernengo, M. G. et al. Prognostic factors in Sézary syndrome: a multivariate analysis of clinical, hematological and immunological features. Ann. Oncol. 9, 857–863 (1998).
Kirsch, I. R. et al. TCR sequencing facilitates diagnosis and identifies mature T cells as the cell of origin in CTCL. Sci. Transl. Med. 7, 308ra158 (2015).
Bolden, J. E., Peart, M. J. & Johnstone, R. W. Anticancer activities of histone deacetylase inhibitors. Nat. Rev. Drug Discov. 5, 769–784 (2006).
Qu, K. et al. Chromatin accessibility landscape of cutaneous T cell lymphoma and dynamic response to HDAC inhibitors. Cancer Cell 32, 27–41.e4 (2017).
Regev, A. et al. The Human Cell Atlas. Elife 6, e27041 (2017).
Birnbaum, M. E. et al. Deconstructing the peptide–MHC specificity of T cell recognition. Cell 157, 1073–1087 (2014).
Newell, E. W. & Davis, M. M. Beyond model antigens: high-dimensional methods for the analysis of antigen-specific T cells. Nat. Biotechnol. 32, 149–157 (2014).
Letourneur, F. & Malissen, B. Derivation of a T cell hybridoma variant deprived of functional T cell receptor α- and β-chain transcripts reveals a nonfunctional α-mRNA of BW5147 origin. Eur. J. Immunol. 19, 2269–2274 (1989).
Huse, M. et al. Spatial and temporal dynamics of T cell receptor signaling with a photoactivatable agonist. Immunity 27, 76–88 (2007).
Glanville, J. et al. Identifying specificity groups in the T cell receptor repertoire. Nature 547, 94–98 (2017).
Mathelier, A. et al. JASPAR 2016: a major expansion and update of the open-access database of transcription factor binding profiles. Nucleic Acids Res. 44.D1, D110–D115 (2016).
Jolma, A. et al. DNA-binding specificities of human transcription factors. Cell 152, 327–339 (2013).
Weirauch, M. T. et al. Determination and inference of eukaryotic transcription factor sequence specificity. Cell 158, 1431–1443 (2014).
Mumbach, M. R. et al. Enhancer connectome in primary human cells identifies target genes of disease-associated DNA elements. Nat. Genet. 49, 1602–1612 (2017).
Acknowledgements
We thank members of the Chang, Davis and Greenleaf laboratories, including Y. Shen and K. Qu, for helpful discussions. We thank X. Ji, D. Wagh and J. Coller at the Stanford Functional Genomics Facility. This work was supported by the Parker Institute for Cancer Immunotherapy (A.T.S., H.Y.C. and M.M.D.), the US National Institutes of Health (NIH) grants P50HG007735 (H.Y.C. and W.J.G.), 5U19AI057229 (M.M.D.), and U19AI057266 (W.J.G), and the Scleroderma Research Foundation (H.Y.C.). A.T.S. was supported by a Parker Bridge Scholar Award from the Parker Institute for Cancer Immunotherapy and a Cancer Research Institute Irvington Fellowship from the Cancer Research Institute. N.S. was supported by the National Multiple Sclerosis Society Postdoctoral Fellowship. J.D.B. acknowledges the Broad Institute Fellows and Harvard Society of Fellows programs for funding. M.R.C. was supported by a grant from the Leukemia and Lymphoma Society Career Development Program. W.J.G. is a Chan Zuckerberg Biohub investigator. M.M.D. is an investigator of the Howard Hughes Medical Institute. Sequencing was performed by the Stanford Functional Genomics Facility (which is supported by NIH grant S10OD018220).
Author information
Authors and Affiliations
Contributions
A.T.S., N.S., M.M.D. and H.Y.C. conceived the project; A.T.S, N.S. and J.D.B. performed experiments and analyzed data; B.W. and Y.Q. performed T-ATAC-seq experiments; R.L., J.M.G., M.R.M. and D.G.G. performed ensemble ATAC-seq experiments and analyzed data; W.S.S. performed TCR-seq experiments; Y.W., A.J.R., K.R.P., C.A.L., A.N.S. and M.R.C. analyzed data; M.S.K. and Y.H.K. obtained clinical specimens; H.Y.C, M.M.D, W.J.G. and P.A.K. guided experiments and data analysis; and A.T.S, M.M.D. and H.Y.C. wrote the manuscript with input from all of the authors.
Corresponding authors
Ethics declarations
Competing interests
H.Y.C. and W.J.G. are founders of Epinomics and members of its scientific advisory board. H.Y.C. is a founder of Accent Therapeutics and a member of its scientific advisory board. H.Y.C. is a member of the scientific advisory board of Spring Discovery.
Additional information
Publisher’s note: Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Supplementary information
Supplementary Figures
Supplementary Figures 1–8
Rights and permissions
About this article
Cite this article
Satpathy, A.T., Saligrama, N., Buenrostro, J.D. et al. Transcript-indexed ATAC-seq for precision immune profiling. Nat Med 24, 580–590 (2018). https://doi.org/10.1038/s41591-018-0008-8
Received:
Accepted:
Published:
Issue Date:
DOI: https://doi.org/10.1038/s41591-018-0008-8
This article is cited by
-
Single-cell technologies in multiple myeloma: new insights into disease pathogenesis and translational implications
Biomarker Research (2023)
-
Epigenetic remodeling of the immune landscape in cancer: therapeutic hurdles and opportunities
Journal of Biomedical Science (2023)
-
The technological landscape and applications of single-cell multi-omics
Nature Reviews Molecular Cell Biology (2023)
-
Profiling the epigenetic landscape of the antigen receptor repertoire: the missing epi-immunogenomics data
Nature Methods (2023)
-
Single-cell RNA sequencing to identify cellular heterogeneity and targets in cardiovascular diseases: from bench to bedside
Basic Research in Cardiology (2023)