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.

  • Technical Report
  • Published:

Profiling epigenetic age in single cells

Abstract

DNA methylation dynamics have emerged as a promising biomarker of mammalian aging, with multivariate machine learning models (‘epigenetic clocks’) enabling measurement of biological age in bulk tissue samples. However, intrinsically sparse and binarized methylation profiles of individual cells have so far precluded the assessment of aging in single-cell data. Here we introduce scAge, a statistical framework for epigenetic age profiling at single-cell resolution, and validate our approach in mice. Our method recapitulates the chronological age of tissues while uncovering heterogeneity among cells. We show accurate tracking of the aging process in hepatocytes, demonstrate attenuated epigenetic aging in muscle stem cells and track age dynamics in embryonic stem cells. We also use scAge to reveal, at the single-cell level, a natural and stratified rejuvenation event occurring during early embryogenesis. We provide our framework as a resource to enable exploration of epigenetic aging trajectories at single-cell resolution.

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: Designing the scAge framework.
Fig. 2: scAge tracks aging in hepatocytes and embryonic fibroblasts.
Fig. 3: MuSCs display attenuated epigenetic aging.
Fig. 4: Culture conditions influence epigenetic age in single ESCs.
Fig. 5: An epigenetic rejuvenation event during mouse embryogenesis.
Fig. 6: Lineage-specific resolution reveals stratification in the epigenetic rejuvenation event.

Similar content being viewed by others

Data availability

All data used in this work were obtained from publicly available repositories. Processed single-cell coverage matrices were downloaded from GEO under the following accession nos.: GSE68642 (ref. 27), GSE121436 (ref. 38), GSE56879 (ref. 25) and GSE121690 (ref. 28). Trimmed sequencing files for the hepatocyte/MEF study were downloaded from the SRA under accession no. SRA344045 (ref. 23). Bulk processed methylation data used for model training were downloaded from GEO under accession no. GSE120132 (ref. 34).

Code availability

The scAge framework is publicly available at https://github.com/alex-trapp/scAge.

References

  1. Horvath, S. & Raj, K. DNA methylation-based biomarkers and the epigenetic clock theory of ageing. Nat. Rev. Genet. 19, 371–384 (2018).

    Article  CAS  PubMed  Google Scholar 

  2. López-Otín, C., Blasco, M. A., Partridge, L., Serrano, M. & Kroemer, G. The hallmarks of aging. Cell 153, 1194–1217 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  3. Han, Y. et al. New targeted approaches for epigenetic age predictions. BMC Biol. 18, 71 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Horvath, S. DNA methylation age of human tissues and cell types. Genome Biol. 14, R115 (2013).

  5. Lister, R. et al. Human DNA methylomes at base resolution show widespread epigenomic differences. Nature 462, 315–322 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Meissner, A. et al. Reduced representation bisulfite sequencing for comparative high-resolution DNA methylation analysis. Nucleic Acids Res. 33, 5868–5877 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Han, Y. et al. Targeted methods for epigenetic age predictions in mice. Sci. Rep. 10, 22439 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Bocklandt, S. et al. Epigenetic predictor of age. PLoS ONE 6, e14821 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Han, Y. et al. Epigenetic age-predictor for mice based on three CpG sites. eLife 7, e37462 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  10. Hannum, G. et al. Genome-wide methylation profiles reveal quantitative views of human aging rates. Mol. Cell 49, 359–367 (2013).

    Article  CAS  PubMed  Google Scholar 

  11. Petkovich, D. A. et al. Using DNA methylation profiling to evaluate biological age and longevity interventions. Cell Metab. 25, 954–960 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Meer, M. V., Podolskiy, D. I., Tyshkovskiy, A. & Gladyshev, V. N. A whole lifespan mouse multi-tissue DNA methylation clock. eLife 7, e40675 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  13. Levine, M. E. et al. An epigenetic biomarker of aging for lifespan and healthspan. Aging 10, 573–591 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  14. Lu, A. T. et al. DNA methylation GrimAge strongly predicts lifespan and healthspan. Aging 11, 303–327 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Belsky, D. W. et al. Quantification of the pace of biological aging in humans through a blood test, the DunedinPoAm DNA methylation algorithm. eLife 9, e54870 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Mammalian Methylation Consortium. Universal DNA methylation age across mammalian tissues. Preprint at bioRxiv https://doi.org/10.1101/2021.01.18.426733 (2021).

  17. Lu, Y. et al. Reprogramming to recover youthful epigenetic information and restore vision. Nature 588, 124–129 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Gill, D. et al. Multi-omic rejuvenation of human cells by maturation phase transient reprogramming. Preprint at bioRxiv https://doi.org/10.1101/2021.01.15.426786 (2021).

  19. Bell, C. G. et al. DNA methylation aging clocks: challenges and recommendations. Genome Biol. 20, 249 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  20. Olova, N., Simpson, D. J., Marioni, R. E. & Chandra, T. Partial reprogramming induces a steady decline in epigenetic age before loss of somatic identity. Aging Cell 18, e12877 (2019).

    Article  PubMed  Google Scholar 

  21. Sarkar, T. J. et al. Transient non-integrative expression of nuclear reprogramming factors promotes multifaceted amelioration of aging in human cells. Nat. Commun. 11, 1545 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Karemaker, I. D. & Vermeulen, M. Single-cell DNA methylation profiling: technologies and biological applications. Trends Biotechnol. 36, 952–965 (2018).

    Article  CAS  PubMed  Google Scholar 

  23. Gravina, S., Dong, X., Yu, B. & Vijg, J. Single-cell genome-wide bisulfite sequencing uncovers extensive heterogeneity in the mouse liver methylome. Genome Biol. 17, 150 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  24. Almanzar, N. et al. A single-cell transcriptomic atlas characterizes ageing tissues in the mouse. Nature 583, 590–595 (2020).

    Article  CAS  Google Scholar 

  25. Smallwood, S. A. et al. Single-cell genome-wide bisulfite sequencing for assessing epigenetic heterogeneity. Nat. Methods 11, 817–820 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. 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).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Angermueller, C. et al. Parallel single-cell sequencing links transcriptional and epigenetic heterogeneity. Nat. Methods 13, 229–232 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Argelaguet, R. et al. Multi-omics profiling of mouse gastrulation at single-cell resolution. Nature 576, 487–491 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Clark, S. J. et al. scNMT-seq enables joint profiling of chromatin accessibility DNA methylation and transcription in single cells. Nat. Commun. 9, 781 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  30. Argelaguet, R. et al. MOFA+: a statistical framework for comprehensive integration of multi-modal single-cell data. Genome Biol. 21, 111 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  31. Angermueller, C., Lee, H. J., Reik, W. & Stegle, O. DeepCpG: accurate prediction of single-cell DNA methylation states using deep learning. Genome Biol. 18, 67 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  32. Kapourani, C.-A. & Sanguinetti, G. Melissa: Bayesian clustering and imputation of single-cell methylomes. Genome Biol. 20, 61 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  33. Stubbs, T. M. et al. Multi-tissue DNA methylation age predictor in mouse. Genome Biol. 18, 68 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  34. Thompson, M. J. et al. A multi-tissue full lifespan epigenetic clock for mice. Aging 10, 2832–2854 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Zou, H. & Hastie, T. Regularization and variable selection via the elastic net. J. R. Stat. Soc. Ser. B Stat. Methodol. 67, 301–320 (2005).

    Article  Google Scholar 

  36. Xie, W., Baylin, S. B. & Easwaran, H. DNA methylation in senescence, aging and cancer. Oncoscience 6, 291–293 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  37. Wang, T. et al. Epigenetic aging signatures in mice livers are slowed by dwarfism, calorie restriction and rapamycin treatment. Genome Biol. 18, 57 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  38. Hernando-Herraez, I. et al. Ageing affects DNA methylation drift and transcriptional cell-to-cell variability in mouse muscle stem cells. Nat. Commun. 10, 4361 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  39. García-Prat, L. et al. Autophagy maintains stemness by preventing senescence. Nature 529, 37–42 (2016).

    Article  PubMed  Google Scholar 

  40. Schwörer, S. et al. Epigenetic stress responses induce muscle stem-cell ageing by Hoxa9 developmental signals. Nature 540, 428–432 (2016).

    Article  PubMed  Google Scholar 

  41. Novak, J. S. et al. Human muscle stem cells are refractory to aging. Aging Cell 20, e13411 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Kerepesi, C., Zhang, B., Lee, S.-G., Trapp, A. & Gladyshev, V. N. Epigenetic clocks reveal a rejuvenation event during embryogenesis followed by aging. Sci. Adv. 7, eabg6082 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Ficz, G. et al. FGF signaling inhibition in ESCs drives rapid genome-wide demethylation to the epigenetic ground state of pluripotency. Cell Stem Cell 13, 351–359 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Gladyshev, V. N. The ground zero of organismal life and aging. Trends Mol. Med. 27, 11–19 (2021).

    Article  CAS  PubMed  Google Scholar 

  45. Pijuan-Sala, B. et al. A single-cell molecular map of mouse gastrulation and early organogenesis. Nature 566, 490–495 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Titus, A. J., Gallimore, R. M., Salas, L. A. & Christensen, B. C. Cell-type deconvolution from DNA methylation: a review of recent applications. Hum. Mol. Genet. 26, R216–R224 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Houseman, E. A. et al. Reference-free deconvolution of DNA methylation data and mediation by cell composition effects. BMC Bioinformatics 17, 259 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  48. Schaum, N. et al. Ageing hallmarks exhibit organ-specific temporal signatures. Nature 583, 596–602 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Rando, T. A. & Wyss-Coray, T. Asynchronous, contagious and digital aging. Nat. Aging 1, 29–35 (2021).

    Article  PubMed  PubMed Central  Google Scholar 

  50. Krueger, F. & Andrews, S. R. Bismark: a flexible aligner and methylation caller for Bisulfite-Seq applications. Bioinformatics 27, 1571–1572 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank T. Fox and A. Ganguly for help with schematic figures. We thank M. Mariotti, A. Shindyapina, S. H. Yim, S.-G. Lee, D. Santesmasses, P. Griffin and Y. Hu for helpful discussions. This work was supported by NIA grants to V.N.G. The funders had no role in study design, data collection and analysis, decision to publish or preparation of the manuscript. Some figures were created with BioRender.com.

Author information

Authors and Affiliations

Authors

Contributions

A.T. and C.K. conceptualized the single-cell age prediction algorithm and processed the training data. A.T. handled all single-cell data, developed the scAge framework and performed all analyses in the manuscript. A.T. and V.N.G. wrote and revised the manuscript with help from C.K. A.T. and V.N.G. conceived the study and V.N.G. supervised the work.

Corresponding author

Correspondence to Vadim N. Gladyshev.

Ethics declarations

Competing interests

Brigham and Women’s Hospital is the sole owner of a provisional patent application directed at this invention in which all authors are named inventors.

Additional information

Peer review information Nature Aging thanks Alex Zhavoronkov, Danica Chen and Lenhard Rudolph for their contribution to the peer review of this work.

Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Extended data figures and tables

Extended Data Fig. 1 Bulk training data characteristics and dimensionality reduction.

a) Age distributions for bulk training data in liver (n = 29), blood (n = 50), muscle (n = 24), and multi-tissue (n = 196) datasets, stratified by gender (female, blue; male, orange). b) Principal component analyses (PCA) across 748,955 CpG sites in liver, blood, muscle, and multi-tissue methylation matrices. For single-tissue datasets, black circles encompass samples that were retained for linear model training to exclude outliers and improve model accuracy. The number of samples before and after filtration is shown in the bottom right of each panel. Color scales depict the age in months of the animal (from young, yellow to old, purple). c) Age distribution in the multi-tissue dataset (n = 196), stratified by tissue type (blood, red; liver, orange; muscle, green; kidney, brown; adipose, yellow; lung, pink).

Extended Data Fig. 2 Relationship between age and bulk methylation level in age-associated CpG sites in liver.

a) Kernel density estimation plots for the top 5 positively and negatively age-correlated CpG sites in the bulk liver data (based on n = 29 samples). CpG genomic positions are shown above each panel, along with the Pearson correlation coefficient (r) between methylation level and age. Colors correspond to the ages of mice (2 m, blue; 10 m, orange; 20 m, red). b) Representative scatterplots showing the relationship between age and methylation level in strongly positively (left) and negatively (right) age-associated CpG sites. Jitter was applied to the x-axis (age) purely for visualization purposes. Regression lines (grey) with 95% confidence intervals (light grey) are shown. Pearson correlation coefficients (r) and associated p-values (p) are shown. Two-tailed Pearson correlation analysis was employed for statistical testing, with statistics for each model treated independently without correction. Colors correspond to the ages of mice (2 m, blue; 10 m, orange; 20 m, red).

Extended Data Fig. 3 Global methylation, coverage, and scDNAm predictions in embryonic fibroblasts and hepatocytes with outliers.

a) Bar plot of mean global methylation (top) and CpG coverage (bottom) in single mouse embryonic fibroblasts and hepatocytes. Each bar represents one cell. MEFs are shown in green, young hepatocytes in blue, and old hepatocytes in red. b) Predicted epigenetic age versus chronological age (top) in all young hepatocytes (blue, n = 11) and old hepatocytes (red, n = 10) across liver (left), multi-tissue (middle) and blood (right) models. Jitter was applied to x-axis (chronological age) strictly for visualization purposes. Pearson correlation (r), the associated p-value (p) and the median absolute error (MedAE) are shown. Two-tailed Pearson correlation analysis was employed for statistical testing with statistics for each model treated independently without correction. Violin plots show kernel density estimation of the data, with the median displayed by a black line. Further analysis of outliers is shown in Extended Data Fig. 5. Dots depict individual cells. c) Predicted epigenetic age, grouped by cell type, across liver (left), multi-tissue (middle) and blood (right) models for MEFs (n = 5, green), young hepatocytes (n = 11, blue), and old hepatocytes (n = 10, red). Two-tailed Welch’s t-test was used for statistical testing, and Bonferroni correction was applied to correct for multiple testing. Box plots show median levels and the first and third quartile, whiskers show up to 1.5× the interquartile range. Dots depict individual cells.

Extended Data Fig. 4 Likelihood distributions in young and old hepatocytes.

Likelihood distributions for all young (blue, n = 11) and old (red, n = 10) hepatocytes, based on scDNAm results from the liver model sampling the top 1% age-associated CpGs per cell. Black lines indicate age of maximum likelihood (predicted epigenetic age), which is depicted numerically in the top right or left corners of each panel. Labels indicate cell identifier, as given in the study metadata on the SRA. Likelihood was calculated by taking the exponential of the log-likelihood profiles, which was subsequently scaled between 0 and 1 to normalize distributions between cells.

Extended Data Fig. 5 Outlier analysis and lack of relationship between scDNAm age and technical covariates in hepatocytes and fibroblasts.

a) Scatterplot depicting the strongly linear relationship between CpG coverage in a single cell (x-axis) and the number of CpGs intersecting with the liver training dataset (y-axis) for embryonic fibroblasts (green, n = 5), young hepatocytes (blue, n = 11) and old hepatocytes (red, n = 10). Regression line (grey) is shown with a 95% confidence interval (light grey). Outlier samples based on scDNAm predictions and PCA analysis in the original study are within the black circle, highlighting these cells are not outliers in regard to CpG coverage. Pearson correlation coefficient (r) and the associated two-tailed p-value (p) are shown. b) Mean global methylation of embryonic fibroblasts (green, n = 5), young (blue, n = 11) and old hepatocytes (red, n = 10). Outlier samples detected during dimensionality reduction and age predictions are circled in black. c, d) Scatterplots depicting the relationship of mean global methylation (left) and CpG coverage (right) with predicted epigenetic age (scDNAm age) for single embryonic fibroblasts (n = 5, green) and hepatocytes (young, n = 11, blue; old, n = 10, red) across liver and multi-tissue datasets with the two outliers included (c) and with the two outliers excluded (d). Regression lines (grey) are shown with a 95% confidence interval (light grey). Two-tailed Pearson correlation analysis was used for statistical testing, with each analysis treated independently without correction. Pearson correlation coefficients (r) and associated two-tailed p-values (p) are shown. No significant relationship is observed in any comparison. The legend in panel (a) applies to all of the panels in this figure.

Extended Data Fig. 6 Pearson correlation distributions and predicted ages in single cells based on various CpG selection parameters.

a) Violin plots depicting the distribution of the Pearson correlation coefficient of scAge-chosen CpGs in embryonic fibroblasts (n = 5, green) and hepatocytes (young, n = 11, blue; old, n = 10, red) based on the selection method. On the left, a percentile-based method is employed, wherein the top x% absolute age-associated CpGs are chosen in every cell. On the right, a defined number of CpGs is chosen across every cell, leading to more uneven distributions due to differential cell CpG coverage. Various parameters for both methods (grey boxes) and their effects on the distributions are shown. Violin plots depict kernel density estimations of the data. Inner boxplots depict median levels (white dot) and first and third quartiles, with whiskers extending up to 1.5x the interquartile range. The central legend applies to all subpanels in this panel. b) Predicted epigenetic ages using the liver model for all embryonic fibroblasts (n = 5) and hepatocytes (young, n = 11; old, n = 10), based on the selection method (left, percentile; right, defined number of CpGs) and parameter. Colors depict the % or number of CpGs chosen for scAge computations (top 0.5% or 100 CpGs, blue; top 1% or 500 CpGs, orange; top 5% or 1,000 CpGs, green; top 10% or 5,000 CpGs, red).

Extended Data Fig. 7 Single-cell epigenetic age predictions differ based on selection mode and training dataset.

Predicted epigenetic ages in all embryonic fibroblasts (green, n = 5), young hepatocytes (blue, n = 11) and old hepatocytes (red, n = 10) using the liver model (left two columns) and multi-tissue models (right two columns) across different CpG selection modes and parameters. Parameters are labeled in grey boxes above the plots. Bonferroni corrections were applied to account for multiple testing. Pearson correlation (r), its associated p-value (p), and the median absolute error (MedAE) are shown for each panel. Two-tailed Pearson correlation analysis was employed for statistical testing. Dashed lines represent the chronological age of animals from which hepatocytes were obtained (4-months-old, dark blue; 26-months-old, dark red). Boxplots depict median levels and the first/third quartile, with whiskers extending up to 1.5x the interquartile range. Individual cells are depicted as points.

Extended Data Fig. 8 Distribution of Pearson correlation coefficients and linear association metrics across training datasets.

a, b) Kernel density estimation plots for (a) Pearson correlation coefficients and (b) linear regression coefficients in processed training reference data for liver (orange), blood (red), muscle (green), and multi-tissue (magenta) datasets. Individual distributions are labeled on the upper right side to indicate which tissue is depicted.

Extended Data Fig. 9 Single-cell coverage distributions and the effect of coverage on scAge predictions.

a) Distributions of single-cell CpG coverage across all 5 datasets analyzed in this study. Dotted lines represent the cutoff value that was used for downstream analysis (at least 500,000 CpGs per cell), in line with previous work38. Given the low sample size and relatively high coverage, no filtration was applied to cells from the Gravina et al. study23. The number of cells passing the filtration cutoff in each dataset is further detailed in Supplementary Table 1. Violin plots depict the kernel density estimation of the data. Inner boxplots depict the median (white dot), as well as the first/third quartile (grey box), with whiskers extending up to 1.5x the interquartile range. Individual dots depict single cells. Colors align with those presented in main figures (Gravina et al: MEFs in green; young hepatocytes in blue; old hepatocytes in red; Hernando-Herraez et al: young MuSCs in blue, old MuSCs in red; Angermueller et al and Smallwood et al: 2i ESCs in yellow, serum ESCs in pink; Argelaguet et al: E4.5 cells in purple, E5.5 cells in dark blue, E6.5 cells in dark green, E7.5 cells in light green). b) Scatterplot depicting the relationship between CpG coverage and predicted epigenetic ages in all unfiltered muscle stem cells38 (n = 275). Dotted black line represents the cutoff of 500,000 CpGs per cell, after which predictions greatly stabilize. MuSCs from young animals are shown in blue, and those from old animals are shown in red.

Extended Data Fig. 10 Single-cell profile simulations and epigenetic age predictions.

a) Heatmap of methylation values in bulk and simulated single-cell profiles. 100 CpGs were randomly selected from a bulk liver sample34, and random Bernoulli distributions were used to generate 10 simulated binary profiles per bulk sample. As the bulk methylation level (top) of CpGs increases from left to right, more simulated single-cell profiles are methylated as opposed to unmethylated. Color scale depicts methylation level from unmethylated (0, black) to methylated (1, white). b) Mean global methylation of bulk samples (blue) and 10 simulated full binary profiles per sample (orange) across 29 bulk liver RRBS samples, arranged from young (left) to old (right). Simulated binary profiles cluster with their bulk source, despite shifting from a fractional to a binary data modality. c, d) Predicted epigenetic age for each simulated binary profile with (c) full coverage of 748,955 CpGs per simulated profile and (d) randomly 10x downsampled coverage of 74,896 distinct CpGs per simulated profile. Profiles are arranged from young (left) to old (right). Age of the animals is denoted by the color of the points (2 m, light green; 10 m, dark green; 20 m, dark blue). Two-tailed Pearson correlation analysis was employed for statistical testing, with statistics for each simulation treated independently without correction. The Pearson correlation coefficient (r), the associated two-tailed p-value (p), median absolute error (MedAE) and mean of the standard deviations for each sample (μ(σ)) are shown. Violin plots in (c) and (d) depict the kernel density estimation of the data. Inner boxplots depict the median (white dot), as well as the first/third quartile (grey box), with whiskers extending up to 1.5x the interquartile range. Individual dots depict simulated single cell profiles.

Supplementary information

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Trapp, A., Kerepesi, C. & Gladyshev, V.N. Profiling epigenetic age in single cells. Nat Aging 1, 1189–1201 (2021). https://doi.org/10.1038/s43587-021-00134-3

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s43587-021-00134-3

This article is cited by

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