Abstract
Natural sequence variation within mitochondrial DNA (mtDNA) contributes to human phenotypes and may serve as natural genetic markers in human cells for clonal and lineage tracing. We recently developed a single-cell multi-omic approach, called ‘mitochondrial single-cell assay for transposase-accessible chromatin with sequencing’ (mtscATAC-seq), enabling concomitant high-throughput mtDNA genotyping and accessible chromatin profiling. Specifically, our technique allows the mitochondrial genome-wide inference of mtDNA variant heteroplasmy along with information on cell state and accessible chromatin variation in individual cells. Leveraging somatic mtDNA mutations, our method further enables inference of clonal relationships among native ex vivo-derived human cells not amenable to genetic engineering-based clonal tracing approaches. Here, we provide a step-by-step protocol for the use of mtscATAC-seq, including various cell-processing and flow cytometry workflows, by using primary hematopoietic cells, subsequent single-cell genomic library preparation and sequencing that collectively take ~3–4 days to complete. We discuss experimental and computational data quality control metrics and considerations for the extension to other mammalian tissues. Overall, mtscATAC-seq provides a broadly applicable platform to map clonal relationships between cells in human tissues, investigate fundamental aspects of mitochondrial genetics and enable additional modes of multi-omic discovery.
This is a preview of subscription content, access via your institution
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
Get just this article for as long as you need it
$39.95
Prices may be subject to local taxes which are calculated during checkout







Data availability
Raw mtscATAC-seq data for the demonstration of the analysis was downloaded from https://www.ncbi.nlm.nih.gov/geo/query/acc.cgi?acc=GSM4472967 and processed with CellRanger v2 to the hg38 reference genome before being processed with mgatk v0.6.1 The comparison single-nucleus ATAC-seq dataset is available at https://www.10xgenomics.com/resources/datasets/5-k-peripheral-blood-mononuclear-cells-pbm-cs-from-a-healthy-donor-next-gem-v-1-1-1-1-standard-2-0-0.
Code availability
A code resource for performing mtscATAC-seq analyses and reproducing the analysis figures in this work is available at https://github.com/caleblareau/mtscatac_protocol. The mgatk package is made freely available and is actively maintained at https://github.com/caleblareau/mgatk. Additional support for interactive variant calling in the R environment has been incorporated in the CRAN Signac package for single-cell chromatin analyses49.
References
Baron, C. S. & van Oudenaarden, A. Unravelling cellular relationships during development and regeneration using genetic lineage tracing. Nat. Rev. Mol. Cell Biol. 20, 753–765 (2019).
VanHorn, S. & Morris, S. A. Next-generation lineage tracing and fate mapping to interrogate development. Dev. Cell 56, 7–21 (2021).
Wagner, D. E. & Klein, A. M. Lineage tracing meets single-cell omics: opportunities and challenges. Nat. Rev. Genet. 21, 410–427 (2020).
Woodworth, M. B., Girskis, K. M. & Walsh, C. A. Building a lineage from single cells: genetic techniques for cell lineage tracking. Nat. Rev. Genet. 18, 230–244 (2017).
Biasco, L. et al. In vivo tracking of human hematopoiesis reveals patterns of clonal dynamics during early and steady-state reconstitution phases. Cell Stem Cell 19, 107–119 (2016).
Ferrari, S. et al. Efficient gene editing of human long-term hematopoietic stem cells validated by clonal tracking. Nat. Biotechnol. 38, 1298–1308 (2020).
Park, S. et al. Clonal dynamics in early human embryogenesis inferred from somatic mutation. Nature 597, 393–397 (2021).
Coorens, T. H. H. et al. Extensive phylogenies of human development inferred from somatic mutations. Nature 597, 387–392 (2021).
Lodato, M. A. et al. Somatic mutation in single human neurons tracks developmental and transcriptional history. Science 350, 94–98 (2015).
Lee-Six, H. et al. Population dynamics of normal human blood inferred from somatic mutations. Nature 561, 473–478 (2018).
Brunner, S. F. et al. Somatic mutations and clonal dynamics in healthy and cirrhotic human liver. Nature 574, 538–542 (2019).
Macaulay, I. C. et al. G&T-seq: parallel sequencing of single-cell genomes and transcriptomes. Nat. Methods 12, 519–522 (2015).
Ludwig, L. S. et al. Lineage tracing in humans enabled by mitochondrial mutations and single-cell genomics. Cell 176, 1325–1339.e22 (2019).
Xu, J. et al. Single-cell lineage tracing by endogenous mutations enriched in transposase accessible mitochondrial DNA. Elife 8, e45105 (2019).
Lareau, C. A. et al. Massively parallel single-cell mitochondrial DNA genotyping and chromatin profiling. Nat. Biotechnol. 39, 451–461 (2021).
Penter, L. et al. Longitudinal single-cell dynamics of chromatin accessibility and mitochondrial mutations in chronic lymphocytic leukemia mirror disease history. Cancer Discov. 11, 3048–3063 (2021).
Taylor, R. W. et al. Mitochondrial DNA mutations in human colonic crypt stem cells. J. Clin. Invest. 112, 1351–1360 (2003).
Teixeira, V. H. et al. Stochastic homeostasis in human airway epithelium is achieved by neutral competition of basal cell progenitors. Elife 2, e00966 (2013).
Stewart, J. B. & Chinnery, P. F. The dynamics of mitochondrial DNA heteroplasmy: implications for human health and disease. Nat. Rev. Genet. 16, 530–542 (2015).
Kang, E. et al. Age-related accumulation of somatic mitochondrial DNA mutations in adult-derived human iPSCs. Cell Stem Cell 18, 625–636 (2016).
Walker, M. A. et al. Purifying selection against pathogenic mitochondrial DNA in human T cells. N. Engl. J. Med. 383, 1556–1563 (2020).
Corces, M. R. et al. Lineage-specific and single-cell chromatin accessibility charts human hematopoiesis and leukemia evolution. Nat. Genet. 48, 1193–1203 (2016).
Corces, M. R. et al. An improved ATAC-seq protocol reduces background and enables interrogation of frozen tissues. Nat. Methods 14, 959–962 (2017).
Montefiori, L. et al. Reducing mitochondrial reads in ATAC-seq using CRISPR/Cas9. Sci. Rep. 7, 1–9 (2017).
Fiskin, E. et al. Single-cell profiling of proteins and chromatin accessibility using PHAGE-ATAC. Nat. Biotechnol. 40, 374–381 (2022).
Mimitou, E. P. et al. Scalable, multimodal profiling of chromatin accessibility, gene expression and protein levels in single cells. Nat. Biotechnol. 39, 1246–1258 (2021).
Santibanez-Koref, M. et al. Assessing mitochondrial heteroplasmy using next generation sequencing: a note of caution. Mitochondrion 46, 302–306 (2019).
Laricchia, K. M. et al. Mitochondrial DNA variation across 56,434 individuals in gnomAD. Genome Res. 32, 569–582 (2022).
Maude, H. et al. NUMT confounding biases mitochondrial heteroplasmy calls in favor of the reference allele. Front. Cell Dev. Biol. 7, 201 (2019).
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).
Tang, Z. et al. A genetic bottleneck of mitochondrial DNA during human lymphocyte development. Mol. Biol. Evol. 39, msac090 (2022).
Lareau, C. A. et al. Droplet-based combinatorial indexing for massive-scale single-cell chromatin accessibility. Nat. Biotechnol. 37, 916–924 (2019).
Domcke, S. et al. A human cell atlas of fetal chromatin accessibility. Science 370, eaba7612 (2020).
Buenrostro, J. D. et al. Single-cell chromatin accessibility reveals principles of regulatory variation. Nature 523, 486–490 (2015).
Cusanovich, D. A. et al. Multiplex single cell profiling of chromatin accessibility by combinatorial cellular indexing. Science 348, 910–914 (2015).
Basu, U., Bostwick, A. M., Das, K., Dittenhafer-Reed, K. E. & Patel, S. S. Structure, mechanism, and regulation of mitochondrial DNA transcription initiation. J. Biol. Chem. 295, 18406–18425 (2020).
Miller, T. E. et al. Mitochondrial variant enrichment from high-throughput single-cell RNA sequencing resolves clonal populations. Nat. Biotechnol. 40, 1030–1034 (2022).
Mulqueen, R. M. et al. High-content single-cell combinatorial indexing. Nat. Biotechnol. 39, 1574–1580 (2021).
Nam, A. S. et al. Somatic mutations and cell identity linked by Genotyping of Transcriptomes. Nature 571, 355–360 (2019).
Rodriguez-Meira, A. et al. Unravelling intratumoral heterogeneity through high-sensitivity single-cell mutational analysis and parallel RNA sequencing. Mol. Cell 73, 1292–1305.e8 (2019).
Stoeckius, M. et al. Simultaneous epitope and transcriptome measurement in single cells. Nat. Methods 14, 865–868 (2017).
Triska, P. et al. Landscape of germline and somatic mitochondrial DNA mutations in pediatric malignancies. Cancer Res. 79, 1318–1330 (2019).
Slyper, M. et al. A single-cell and single-nucleus RNA-Seq toolbox for fresh and frozen human tumors. Nat. Med. 26, 792–802 (2020).
Qian, J. et al. A pan-cancer blueprint of the heterogeneous tumor microenvironment revealed by single-cell profiling. Cell Res. 30, 745–762 (2020).
Wakiro, I. et al. HTAPP_Dissociation of human ovarian cancer resection to a single-cell suspension for single-cell RNA-seq. Available at https://www.protocols.io/view/htapp-dissociation-of-human-ovarian-cancer-resecti-bhbhj2j6 (2020).
Swanson, E. et al. Simultaneous trimodal single-cell measurement of transcripts, epitopes, and chromatin accessibility using TEA-seq. Elife 10, e63632 (2021).
Chen, X. et al. ATAC-see reveals the accessible genome by transposase-mediated imaging and sequencing. Nat. Methods 13, 1013–1020 (2016).
Stoler, N. & Nekrutenko, A. Sequencing error profiles of Illumina sequencing instruments. NAR Genom. Bioinform. 3, lqab019 (2021).
Stuart, T., Srivastava, A., Madad, S., Lareau, C. A. & Satija, R. Single-cell chromatin state analysis with Signac. Nat. Methods 18, 1333–1341 (2021).
Quinlan, A. R. & Hall, I. M. BEDTools: a flexible suite of utilities for comparing genomic features. Bioinformatics 26, 841–842 (2010).
Li, H. & Durbin, R. Fast and accurate long-read alignment with Burrows-Wheeler transform. Bioinformatics 26, 589–595 (2010).
Granja, J. M. et al. ArchR is a scalable software package for integrative single-cell chromatin accessibility analysis. Nat. Genet. 53, 403–411 (2021).
Van der Auwera, G. A. et al. From FastQ data to high confidence variant calls: the Genome Analysis Toolkit best practices pipeline. Curr. Protoc. Bioinform. 43, 11.10.1–11.10.33 (2013).
Acknowledgements
We are grateful to members of the Satpathy, Regev, Sankaran and Ludwig laboratories for helpful discussions. This research was supported by a Stanford Science Fellowship (to C.A.L.) and a Parker Institute of Cancer Immunotherapy Scholarship (to C.A.L.), a Stanford Artificial Intelligence in Medicine and Imaging seed grant (to C.A.L.), a BroadIgnite Award (L.S.L.), R01 DK103794 (to V.G.S.), R33 HL120791 (to V.G.S.), a gift from the Lodish Family to Boston Children’s Hospital (to V.G.S.), the New York Stem Cell Foundation (NYSCF; to V.G.S.) and the Howard Hughes Medical Institute and Klarman Cell Observatory (to A.R.). V.G.S. is an NYSCF-Robertson Investigator. P.K. is an Associated Fellow of the Hector Fellow Academy. L.N. is funded by a fellowship from the MDC-NYU exchange program and is an Associated Fellow of the Hector Fellow Academy. L.S.L. is supported by the Berlin Institute of Health, an Emmy Noether fellowship by the German Research Foundation (DFG, LU 2336/2-1) and a Hector Research Career Development Award. A.T.S. is supported by the National Institutes of Health (NIH) U01CA260852, the Parker Institute for Cancer Immunotherapy, a Career Award for Medical Scientists from the Burroughs Wellcome Fund and a Pew-Stewart Scholars for Cancer Research Award. A.T.S., C.A.L. and L.S.L. are supported by NIH UM1HG012076.
Author information
Authors and Affiliations
Contributions
C.A.L., L.S.L. and C.M. conceived and designed the protocol with supervision from A.R. and V.G.S. K.S. designed and optimized the solid tissue dissociation protocol. V.L. and J.C.G. refined the computational workflow with supervision from C.A.L. and A.T.S. L.N., P.K., K.S., Y.Y. and K.P. independently executed the protocol and provided critical feedback on the manuscript. C.A.L., S.D.P., A.T.S., A.R., V.G.S. and L.S.L. supervised various aspects of this study. All authors contributed to the writing and editing of the protocol.
Corresponding authors
Ethics declarations
Competing interests
The Broad Institute has filed a patent relating to the use of the technology described in this paper for which C.A.L., L.S.L., C.M., A.R. and V.G.S. are named as inventors (US Patent App. 17/251,451). A.R. is a founder and equity holder of Celsius Therapeutics, an equity holder in Immunitas Therapeutics and until 31 August 2020, was a scientific advisory board member of Syros Pharmaceuticals, Neogene Therapeutics, Asimov and Thermo Fisher Scientific. From 1 August 2020, A.R. has been an employee of Genentech. V.G.S. serves as an advisor to and/or has equity in Novartis, Forma, Cellarity, Ensoma and Polaris Partners. A.T.S. is a founder of Immunai and Cartography Biosciences and receives research funding from Merck Research Laboratories and Allogene Therapeutics.
Peer review
Peer review information
Nature Protocols thanks Judith Zaugg and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.
Additional information
Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Related links
Key references using this protocol
Lareau, C. et al. Nat. Biotechnol. 39, 451–461 (2021): https://doi.org/10.1038/s41587-020-0645-6
Walker, M. et al. N. Engl. J. Med. 383, 1556–1563 (2020): https://doi.org/10.1056/NEJMoa2001265
Penter, L. et al. Cancer Discov. 11, 3048–3063 (2021): https://doi.org/10.1158/2159-8290.CD-21-0276
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.
About this article
Cite this article
Lareau, C.A., Liu, V., Muus, C. et al. Mitochondrial single-cell ATAC-seq for high-throughput multi-omic detection of mitochondrial genotypes and chromatin accessibility. Nat Protoc 18, 1416–1440 (2023). https://doi.org/10.1038/s41596-022-00795-3
Received:
Accepted:
Published:
Issue Date:
DOI: https://doi.org/10.1038/s41596-022-00795-3
Comments
By submitting a comment you agree to abide by our Terms and Community Guidelines. If you find something abusive or that does not comply with our terms or guidelines please flag it as inappropriate.