Abstract
In vivo lineage tracing holds great potential to reveal fundamental principles of tissue development and homeostasis. However, current lineage tracing in humans relies on extremely rare somatic mutations, which has limited temporal resolution and lineage accuracy. Here, we developed a generic lineage-tracing tool based on frequent epimutations on DNA methylation, enabled by our computational method MethylTree. Using single-cell genome-wide DNA methylation datasets with known lineage and phenotypic labels, MethylTree reconstructed lineage histories at nearly 100% accuracy across different cell types, developmental stages, and species. We demonstrated the epimutation-based single-cell multi-omic lineage tracing in mouse and human blood, where MethylTree recapitulated the differentiation hierarchy in hematopoiesis. Applying MethylTree to human embryos, we revealed early fate commitment at the four-cell stage. In native mouse blood, we identified ~250 clones of hematopoietic stem cells. MethylTree opens the door for high-resolution, noninvasive and multi-omic lineage tracing in humans and beyond.
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
$32.99 / 30 days
cancel any time
Subscribe to this journal
Receive 12 print issues and online access
$259.00 per year
only $21.58 per issue
Buy this article
- Purchase on SpringerLink
- Instant access to full article PDF
Prices may be subject to local taxes which are calculated during checkout






Similar content being viewed by others
Data availability
The sequencing data for human blood has been submitted to the Genome Sequence Archive database under the accession number HRA008624. Other sequencing data generated in this study have been submitted to NCBI GEO, with the accession number GSE262580. The methylation rate matrix associated with selected genomic regions for each analyzed dataset in our paper, along with each sample metadata and processed human blood dataset,is available via figshare at https://doi.org/10.6084/m9.figshare.27288630 (ref. 78). The accession number and analysis parameters for each analyzed dataset in this study are available in Supplementary Table 1.
Code availability
Scripts for data preprocessing are available at https://github.com/ShouWenWang-Lab/Preprocessing. MethylTree code is available at https://github.com/ShouWenWang-Lab/MethylTree. To reproduce our analysis, please check out our jupyter notebooks at https://github.com/ShouWenWang-Lab/MethylTree_notebooks. A web portal of MethylTree analysis is available at https://wangshouwen.lab.westlake.edu.cn/app/methylserver.
References
Kretzschmar, K. & Watt, F. M. Lineage tracing. Cell 148, 33–45 (2012).
Sun, J. et al. Clonal dynamics of native haematopoiesis. Nature 514, 322–327 (2014).
Pei, W. et al. Polylox barcoding reveals haematopoietic stem cell fates realized in vivo. Nature 548, 456–460 (2017).
McKenna, A. et al. Whole-organism lineage tracing by combinatorial and cumulative genome editing. Science 353, aaf7907 (2016).
Alemany, A., Florescu, M., Baron, C. S., Peterson-Maduro, J. & van Oudenaarden, A. Whole-organism clone tracing using single-cell sequencing. Nature 556, 108–112 (2018).
Bowling, S. et al. An engineered CRISPR-Cas9 mouse line for simultaneous readout of lineage histories and gene expression profiles in single cells. Cell 181, 1410–1422.e27 (2020).
Chan, M. M. et al. Molecular recording of mammalian embryogenesis. Nature 570, 77–82 (2019).
Kalhor, R. et al. Developmental barcoding of whole mouse via homing CRISPR. Science 361, eaat9804 (2018).
Xie, L. et al. Comprehensive spatiotemporal mapping of single-cell lineages in developing mouse brain by CRISPR-based barcoding. Nat. Methods https://doi.org/10.1038/s41592-023-01947-3 (2023).
Liu, K. et al. Mapping single-cell-resolution cell phylogeny reveals cell population dynamics during organ development. Nat. Methods https://doi.org/10.1038/s41592-021-01325-x (2021).
Choi, J. et al. A time-resolved, multi-symbol molecular recorder via sequential genome editing. Nature 608, 98–107 (2022).
Wagner, D. E. & Klein, A. M. Lineage tracing meets single-cell omics: opportunities and challenges. Nat. Rev. Genet. https://doi.org/10.1038/s41576-020-0223-2 (2020).
Li, L. et al. A mouse model with high clonal barcode diversity for joint lineage, transcriptomic, and epigenomic profiling in single cells. Cell https://doi.org/10.1016/j.cell.2023.09.019 (2023).
Weinreb, C., Rodriguez-Fraticelli, A., Camargo, F. D. & Klein, A. M. Lineage tracing on transcriptional landscapes links state to fate during differentiation. Science 367, eaaw3381 (2020).
Rodriguez-Fraticelli, A. E. et al. Single-cell lineage tracing unveils a role for TCF15 in haematopoiesis. Nature https://doi.org/10.1038/s41586-020-2503-6 (2020).
Rodriguez-Fraticelli, A. E. et al. Clonal analysis of lineage fate in native haematopoiesis. Nature 553, 212–216 (2018).
Patel, S. H. et al. Lifelong multilineage contribution by embryonic-born blood progenitors. Nature 606, 747–753 (2022).
Jindal, K. et al. Single-cell lineage capture across genomic modalities with CellTag-multi reveals fate-specific gene regulatory changes. Nat. Biotechnol. https://doi.org/10.1038/s41587-023-01931-4 (2023).
Biddy, B. A. et al. Single-cell mapping of lineage and identity in direct reprogramming. Nature 564, 219–224 (2018).
Yang, D. et al. Lineage tracing reveals the phylodynamics, plasticity, and paths of tumor evolution. Cell 185, 1905–1923.e25 (2022).
Quinn, J. J. et al. Single-cell lineages reveal the rates, routes, and drivers of metastasis in cancer xenografts. Science 371, eabc1944 (2021).
Goyal, Y. et al. Diverse clonal fates emerge upon drug treatment of homogeneous cancer cells. Nature https://doi.org/10.1038/s41586-023-06342-8 (2023).
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).
Abyzov, A. & Vaccarino, F. M. Cell lineage tracing and cellular diversity in humans. Annu. Rev. Genomics Hum. Genet. 21, 101–116 (2020).
Mitchell, E. et al. Clonal dynamics of haematopoiesis across the human lifespan. Nature 606, 343–350 (2022).
Fabre, M. A. et al. The longitudinal dynamics and natural history of clonal haematopoiesis. Nature 606, 335–342 (2022).
Ludwig, L. S. et al. Lineage tracing in humans enabled by mitochondrial mutations and single-cell genomics. Cell 176, 1325–1339.e22 (2019).
Campbell, P. et al. Mitochondrial mutation, drift and selection during human development and ageing. Preprint at Research Square https://doi.org/10.21203/rs.3.rs-3083262/v1 (2023).
Wang, X. et al. Clonal expansion dictates the efficacy of mitochondrial lineage tracing in single cells. Preprint at bioRxiv https://doi.org/10.1101/2024.05.15.594338 (2024).
Weng, C. et al. Deciphering cell states and genealogies of human hematopoiesis. Nature https://doi.org/10.1038/s41586-024-07066-z (2024).
Lynch, M. Evolution of the mutation rate. Trends Genet. 26, 345–352 (2010).
Landan, G. et al. Epigenetic polymorphism and the stochastic formation of differentially methylated regions in normal and cancerous tissues. Nat. Genet. 44, 1207–1214 (2012).
Wang, Q. et al. Imprecise DNMT1 activity coupled with neighbor-guided correction enables robust yet flexible epigenetic inheritance. Nat. Genet. 52, 828–839 (2020).
Xie, H. et al. Genome-wide quantitative assessment of variation in DNA methylation patterns. Nucleic Acids Res. 39, 4099–4108 (2011).
Ushijima, T. et al. Fidelity of the methylation pattern and its variation in the genome. Genome Res. 13, 868–874 (2003).
Brocks, D. et al. Intratumor DNA methylation heterogeneity reflects clonal evolution in aggressive prostate cancer. Cell Rep. 8, 798–806 (2014).
Mazor, T. et al. DNA methylation and somatic mutations converge on the cell cycle and define similar evolutionary histories in brain tumors. Cancer Cell 28, 307–317 (2015).
Bian, S. et al. Single-cell multiomics sequencing and analyses of human colorectal cancer. Science 362, 1060–1063 (2018).
Bian, S. et al. Integrative single-cell multiomics analyses dissect molecular signatures of intratumoral heterogeneities and differentiation states of human gastric cancer. Natl Sci. Rev. 10, nwad094 (2023).
Wang, Y. et al. Single-cell dissection of the multiomic landscape of high-grade serous ovarian cancer. Cancer Res. 82, 3903–3916 (2022).
Shipony, Z. et al. Dynamic and static maintenance of epigenetic memory in pluripotent and somatic cells. Nature 513, 115–119 (2014).
Yatabe, Y., Tavaré, S. & Shibata, D. Investigating stem cells in human colon by using methylation patterns. Proc. Natl Acad. Sci. USA 98, 10839–10844 (2001).
Gabbutt, C. et al. Fluctuating methylation clocks for cell lineage tracing at high temporal resolution in human tissues. Nat. Biotechnol. 40, 720–730 (2022).
Gaiti, F. et al. Epigenetic evolution and lineage histories of chronic lymphocytic leukaemia. Nature 569, 576–580 (2019).
Chaligne, R. et al. Epigenetic encoding, heritability and plasticity of glioma transcriptional cell states. Nat. Genet. 53, 1469–1479 (2021).
Liu, Y. et al. Single-cell methylation sequencing data reveal succinct metastatic migration histories and tumor progression models. Genome Res. 33, 1089–1100 (2023).
Grady, W. M. & Carethers, J. M. Genomic and epigenetic instability in colorectal cancer pathogenesis. Gastroenterology 135, 1079–1099 (2008).
Greenberg, M. V. C. & Bourc’his, D. The diverse roles of DNA methylation in mammalian development and disease. Nat. Rev. Mol. Cell Biol. 20, 590–607 (2019).
Smallwood, S. A. et al. Single-cell genome-wide bisulfite sequencing for assessing epigenetic heterogeneity. Nat. Methods 11, 817–820 (2014).
Guo, H. et al. Single-cell methylome landscapes of mouse embryonic stem cells and early embryos analyzed using reduced representation bisulfite sequencing. Genome Res. 23, 2126–2135 (2013).
Liu, H. et al. Single-cell DNA methylome and 3D multi-omic atlas of the adult mouse brain. Nature 624, 366–377 (2023).
Bai, D. et al. Simultaneous single-cell analysis of 5mC and 5hmC with SIMPLE-seq. Nat. Biotechnol. https://doi.org/10.1038/s41587-024-02148-9 (2024).
Adolph, S. C. & Hardin, J. S. Estimating phenotypic correlations: correcting for bias due to intraindividual variability. Funct. Ecol. 21, 178–184 (2007).
Spearman, C. The proof and measurement of association between two things. Am. J. Psychol. 15, 72–101 (1904).
Saitou, N. & Nei, M. The neighbor-joining method: a new method for reconstructing phylogenetic trees. Mol. Biol. Evol. 4, 406–425 (1987).
Lefort, V., Desper, R. & Gascuel, O. FastME 2.0: a comprehensive, accurate, and fast distance-based phylogeny inference program. Mol. Biol. Evol. 32, 2798–2800 (2015).
Li, L. et al. Single-cell RNA-seq analysis maps development of human germline cells and gonadal niche interactions. Cell Stem Cell 20, 858–873.e4 (2017).
Li, L. et al. Dissecting the epigenomic dynamics of human fetal germ cell development at single-cell resolution. Cell Res. https://doi.org/10.1038/s41422-020-00401-9 (2020).
Wang, S.-W., Herriges, M. J., Hurley, K., Kotton, D. N. & Klein, A. M. CoSpar identifies early cell fate biases from single-cell transcriptomic and lineage information. Nat. Biotechnol. https://doi.org/10.1038/s41587-022-01209-1 (2022).
Zhou, W. et al. DNA methylation loss in late-replicating domains is linked to mitotic cell division. Nat. Genet. 50, 591–602 (2018).
Rossant, J. & Tam, P. P. L. Blastocyst lineage formation, early embryonic asymmetries and axis patterning in the mouse. Development 136, 701–713 (2009).
Dietrich, J.-E. & Hiiragi, T. Stochastic patterning in the mouse pre-implantation embryo. Development 134, 4219–4231 (2007).
Goolam, M. et al. Heterogeneity in Oct4 and Sox2 targets biases cell fate in 4-cell mouse embryos. Cell 165, 61–74 (2016).
Wang, J. et al. Asymmetric expression of LincGET biases cell fate in two-cell mouse embryos. Cell 175, 1887–1901.e18 (2018).
Guo, F. et al. Single-cell multi-omics sequencing of mouse early embryos and embryonic stem cells. Cell Res. 27, 967–988 (2017).
Qi, S. et al. Single-cell multiomics analyses of spindle-transferred human embryos suggest a mostly normal embryonic development. PLoS Biol. 20, e3001741 (2022).
Mikkola, H. K. A. & Orkin, S. H. The journey of developing hematopoietic stem cells. Development 133, 3733–3744 (2006).
Rybtsov, S., Ivanovs, A., Zhao, S. & Medvinsky, A. Concealed expansion of immature precursors underpins acute burst of adult HSC activity in foetal liver. Development 143, 1284–1289 (2016).
Henninger, J. et al. Clonal fate mapping quantifies the number of haematopoietic stem cells that arise during development. Nat. Cell Biol. 19, 17–27 (2017).
Ganuza, M. et al. Lifelong haematopoiesis is established by hundreds of precursors throughout mammalian ontogeny. Nat. Cell Biol. 19, 1153–1163 (2017).
Kissa, K. & Herbomel, P. Blood stem cells emerge from aortic endothelium by a novel type of cell transition. Nature 464, 112–115 (2010).
Boisset, J.-C. et al. In vivo imaging of haematopoietic cells emerging from the mouse aortic endothelium. Nature 464, 116–120 (2010).
Luo, C. et al. Single nucleus multi-omics identifies human cortical cell regulatory genome diversity. Cell Genom. 2, 100107 (2022).
Cao, Y. et al. Single-cell bisulfite-free 5mC and 5hmC sequencing with high sensitivity and scalability. Proc. Natl Acad. Sci. USA 120, e2310367120 (2023).
Pei, W. et al. Resolving fates and single-cell transcriptomes of hematopoietic stem cell clones by PolyloxExpress barcoding. Cell Stem Cell 27, 383–395.e8 (2020).
Clark, S. J. et al. Genome-wide base-resolution mapping of DNA methylation in single cells using single-cell bisulfite sequencing (scBS-seq). Nat. Protoc. 12, 534–547 (2017).
Islam, S. et al. Characterization of the single-cell transcriptional landscape by highly multiplex RNA-seq. Genome Res. 21, 1160–1167 (2011).
Chen, M., Fu, R., Chen, Y., Li L. & Wang, S.-W. MethylTree: high-resolution, noninvasive single-cell lineage tracing in mice and humans based on DNA methylation epimutations. figshare https://doi.org/10.6084/m9.figshare.27288630 (2024).
Acknowledgements
We are grateful to D. Li, H. Yu and K. Zhang from Westlake University for their stimulating discussions. We thank W. Liu and D. Ruan from Zhejiang University for sharing LARRY lentivirus. We acknowledge H. Guo from Zhejiang University as well as W. Yue and H. Yan from Beijing Institute of Radiation Medicine for their help. We also thank other laboratory members for their input. L.L. is supported by National Key Research and Development Project of China (2024YFA1306600). We acknowledge support from NSFC (grant no. 32470700), Westlake High-Performance Computing Center, and ‘Pioneer’ and ‘Leading Goose’ R&D Programs of Zhejiang province (grant nos. 2024SSYS0034 and 2024SSYS0034).
Author information
Authors and Affiliations
Contributions
S.-W.W. and L.L. conceived the project. S.-W.W. acquired funding, developed MethylTree, designed experiments, carried out biological applications and wrote the manuscript with input from other authors. M.C. performed all experiments. R.F. carried out data analyses and generated figures. Y.C. developed the web portal. L.L. and S.-W.W. supervised M.C. to set up the experimental system and generate experimental datasets. S.-W.W. supervised the entire project.
Corresponding authors
Ethics declarations
Competing interests
S.-W.W. is named inventor on a patent application for MethylTree (PCT/CN2024/095497). The other authors declare no competing interests.
Peer review
Peer review information
Nature Methods thanks Simon Anders and the other, anonymous, reviewer(s) for their contribution to the peer review of this work. Peer reviewer reports are available. Primary Handling Editor: Lei Tang, in collaboration with the Nature Methods 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 Systematic characterization of MethylTree performance in a homogeneous population.
a, b, Analysis on simulated single-cell expansion with more realistic features. a, The impact of division-free CpG mutations on lineage inference accuracy. After simulated clonal expansion as in Fig. 1d, we randomly mutated a given fraction of CpG sites in each of the 128 cells. b, Heatmap of lineage accuracy as a function of CpG coverage and the variation of epimutation rate controlled by the parameter \(\lambda\). Compared with Fig. 1f, we modeled epimutation on a diploid genome with a CpG-site specific epimutation rate sampled from a uniform distribution with a maximum value \(\lambda\). Each observed CpG status is obtained from sampling once on the same CpG site from either of the two DNA molecules. c–j, MethylTree analysis of a clonal expansion dataset of human HEK 293T cells. c, Heatmap of the similarity matrix computed with the cell-by-CpG matrix, without binning. d, Schematic of region selection. Non-overlapping 500-bp genomic bins with an intermediate methylation rate between \({m}_{0}\) and \({m}_{1}\) were selected. e, Merging neighboring bins after selection in d. This procedure was used in analyzing all datasets in this article. f, Heatmap of MethylTree lineage accuracies on the 293T dataset using ‘merged’ genomic regions selected at different thresholds according to e. The parameters indicated on this plot (m0 = 0.5, m1 = 0.9) were used to generate Fig. 1i–k. g, A scatter plot showing the number of genomic regions associated with each selection and the corresponding accuracy of MethylTree-inferred lineages, using the data from f. The selection parameters (m0, m1) for some data points are highlighted. h, Number of detected CpG sites per cell on the methylation embedding of 293T cells. i, Lineage accuracy using different metrics to compute the cell-cell similarity. With Euclidean distance matrix X, we converted it to a similarity with 1 − X/max(X), where \(\max (X)\) is the largest value in this matrix. j, Similarity heatmap ordered with the phylogenetic tree inferred from the neighbor-joining56 (NJ, left) or FastME57 (right) method.
Extended Data Fig. 2 Lineage inference from human embryonic stem cells and colorectal cancer.
a–c, Lineage analysis of clonal expansion of H9 human embryonic stem cells. a, Schematic of our experimental design, created using BioRender.com. There were five clones generated in this experiment. b, Heatmap of MethylTree lineage accuracies on the H9 dataset, similar with Extended Data Fig. 1f. c, Heatmap of the similarity matrix of the H9 dataset before (left) and after (right) correlation-bias correction. The color bar shows the actual clonal identify of each cell. d–f, Lineage inference from human colorectal cancer. Data is obtained from patient CRC11 in Bian et al.38. d, Schematic of tissue sampling and cell profiling, created using BioRender.com. e, Heatmap of the cell-cell similarity matrix computed from single-cell DNA methylation. Here, A1–A6 and B were inferred cancer lineages based on copy number variations (CNV) in the original analysis by Bian et al. NC marks the normal cells. f, Lineage phylogenetic tree inferred from the methylation matrix. Same color as e.
Extended Data Fig. 3 MethylTree analysis in a heterogeneous population.
a–c, Lineage inference from simulated differentiation. a, Heatmap of lineage accuracy as a function of CpG coverage and the variation of the epimutation rate controlled by \(\lambda\). Here, we simulated differentiation on a haploid genome with a site-specific epimutation rate sampled from a uniform distribution with a maximum value \(\lambda\). Here, the lineage-specific CpG fraction \(\alpha =0.5\). b, Inferred lineage accuracy from simulated differentiation with different fractions of cell-type-specific CpG sites \((1-\alpha )\). The cell-type signals are not removed. c, Lineage accuracy after removing cell-type signals, evaluated at different fractions of lineage-specific CpG sites \((\alpha )\). d, Heatmap of methylation similarity associated with fetus_2 from 17 weeks. e, Inferred lineage tree from d, colored by inferred methyl-clones. f, Methylation embedding colored by methyl-clone ID (top) or FGC sub-types (bottom). g, Heatmap of MethylTree lineage accuracies associated with Fig. 2l using different region choices. The selected regions associated with (0.3,0.6) were re-used to analyze other datasets in Fig. 2 and Extended Data Fig. 3. h, Similarity heatmaps of FGCs and somatic cells from two 7-week human embryos. Left panel: the raw similarity matrix; right panel: after removing cell-type-specific signals.
Extended Data Fig. 4 Analysis of the single-cell multi-omic blood dataset from mouse.
a, Heatmap showing the expression of cell-type-specific marker genes (columns) in each annotated cell types (rows) in Fig. 3c. Expression values were column-wise normalized by the highest value in each column. b, Bar plot of cell counts of each cell type identified in this dataset. c, Histogram of LARRY clone sizes in this dataset. d, Heatmap of MethylTree lineage accuracies associated with different region choices on these blood cells. We highlight the parameters used to generate Fig. 3f. e, Lineage accuracy computed with non-overlapping bins at different sizes, with either correlation-bias correction or not. f, Box plot of lineage accuracies at different genomic coverages. At each coverage, results from all genomic choices are shown. See Fig. 1n for box plot description. g, Heatmap of clonal coupling scores computed from the observed LARRY lineage barcodes. h, Pseudobulk DNA methylation profiles on genomic regions not specifically related to hematopoiesis. Otherwise, same as Fig. 3m. i, Fraction of clone-specific CpG sites in different genomic contexts. These were differentially methylated CpG sites between the two largest clones in this dataset. WCGW: a solo CpG site franked by either A or T; CGI: CpG islands; Prom_CGI: CGI-enriched promoter region (within 2000 bp from transcription starting site); Prom_nonCGI: CGI-depleted promoter region; Genebody: gene body region; LINE: long interspersed nuclear elements; LTR: long terminal repeats. Results from randomly sampled CpG sites are also shown. **, one-sided p-value < 0.01, obtained from directly simulating the null distribution. See Methods.
Extended Data Fig. 5 Analysis of the single-cell multi-omic blood dataset from human.
a, Heatmap showing marker gene expression of each cell type. b, Similarity heatmap created the same as in Fig. 4e, but for all the cells passing methylation quality control. c, Heatmap of MethylTree lineage accuracies associated with different region choices.
Extended Data Fig. 6 MethylTree analysis on developing human and mouse embryos.
a, Heatmap of MethylTree lineage accuracies associated with different region choices on 4-cell-stage cells from mouse embryos. The selected regions associated with (0.4,0.6) were re-used to analyze mouse datasets from other stages in Fig. 5 and this figure. b, Methylation similarity heatmaps of mouse cells from other developmental stages, with the color bar indicating their embryonic origins. c, d, Same as a and b, but for cells from human embryos. The selected regions associated with (0.2,0.5) were re-used to analyze human datasets from other stages in Fig. 5 and this figure. e–g, Methylation similarity heatmaps and reconstructed lineages (with support values from bootstrap sampling) for additional three human embryos, in addition to those shown in Fig. 5h–k. These include E5 from day 5 (e), E7 from day 6 (f), and E8 from day 6 (g).
Extended Data Fig. 7 MethylTree analysis on mouse HSCs.
a, Heatmap of MethylTree lineage accuracies associated with different region choices on HSCs from mouse LL731. We highlight parameters used to generate Fig. 6b, and also the choice used in our previous study13. The same set of genomic regions was re-used in analyzing the remaining HSC datasets in Fig. 6 and this figure. b, Bar plot of adjusted rank index associated with each HSC dataset. c, Bar plot of the fraction of cells among the multi-cell methy-clones. d, HSC clone number inference on mouse LL653E1. From left to right: methylation similarity matrix, inferred lineage tree, distribution of putative clone sizes (same as Fig. 6e), and HSC clone number inference based on the observed singleton cell fraction (same as Fig. 6f). e, HSC clone number inference on mouse LL653E6. Otherwise, same as d.
Extended Data Fig. 8 Summary of all datasets analyzed in this study.
a, Schematic of the global methylation dynamics over the life time of an individual, created using BioRender.com. Our study analyzed datasets across all three key stages of methylation dynamics, including two global de-methylation waves before birth and a stable period after birth. b, Bar plot comparing lineage accuracies from raw similarity or corrected (cell-type-signal removed if needed) similarity across all datasets analyzed in this study. c, Bar plot comparing lineage accuracies from using all 500-bp bins, selected genomic regions, all single-CpG sites, or from a randomized cell ordering, across all datasets analyzed in this study. The accuracies from selected-region and single-CpG methods are significantly higher than those from randomization, each with a p-value < 0.0005.
Extended Data Fig. 9 Comparison between different lineage tracing methods in humans.
We assume that one Gigabyte bases cost 5$ here. This cost could decrease over time as the technology improves.
Supplementary information
Supplementary Table 1
Summary of all the datasets analyzed in this study.
Supplementary Table 2
Primers used in scBS-seq and Camellia-seq.
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
Chen, M., Fu, R., Chen, Y. et al. High-resolution, noninvasive single-cell lineage tracing in mice and humans based on DNA methylation epimutations. Nat Methods 22, 488–498 (2025). https://doi.org/10.1038/s41592-024-02567-1
Received:
Accepted:
Published:
Issue date:
DOI: https://doi.org/10.1038/s41592-024-02567-1
This article is cited by
-
Single-cell lineage tracing techniques in hematology: unraveling the cellular narrative
Journal of Translational Medicine (2025)
-
MOADE: a multimodal autoencoder for dissociating bulk multi-omics data
Genome Biology (2025)
-
Clonal tracing with somatic epimutations reveals dynamics of blood ageing
Nature (2025)