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.

  • Resource
  • Published:

Enhanced epigenetic profiling of classical human monocytes reveals a specific signature of healthy aging in the DNA methylome

Abstract

The impact of healthy aging on molecular programming of immune cells is poorly understood. Here we report comprehensive characterization of healthy aging in human classical monocytes, with a focus on epigenomic, transcriptomic and proteomic alterations, as well as the corresponding proteomic and metabolomic data for plasma, using healthy cohorts of 20 young and 20 older males (~27 and ~64 years old on average). For each individual, we performed enhanced reduced representation bisulfite sequencing-based DNA methylation profiling, which allowed us to identify a set of age-associated differentially methylated regions (DMRs)—a novel, cell-type-specific signature of aging in the DNA methylome. Hypermethylation events were associated with H3K27me3 in the CpG islands near promoters of lowly expressed genes, while hypomethylated DMRs were enriched in H3K4me1-marked regions and associated with age-related increase of expression of the corresponding genes, providing a link between DNA methylation and age-associated transcriptional changes in primary human cells.

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: Integrated multiomics profiling of healthy aging.
Fig. 2: Low-magnitude transcriptional changes and distinct proteome alterations in monocytes during healthy aging.
Fig. 3: RRBS sequencing reveals robust continuous regions of age-associated DNA hyper- and hypomethylation (up and down DMRs).
Fig. 4: Distinct chromatin signatures of up and down age-associated DMRs.
Fig. 5: Distinct influence of up and down DMRs on gene expression.
Fig. 6: Up and down DMRs behave differently in various pathological contexts.
Fig. 7: Down DMRs are enriched in genetic susceptibilities loci for asthma, MS, glycated Hb and total protein.

Similar content being viewed by others

Data availability

Raw human sequencing data (Figs. 24) are deposited in the Synapse repository (https://www.synapse.org/#!Synapse:syn22020090/wiki/602603) and available for download upon request to the corresponding author. Please provide information about the principle investigator (name, affiliation, email address and telephone number) and official email from the institutional signing official. The mass spectrometry proteomics data for monocyte lysates have been deposited to the ProteomeXchange Consortium via the PRIDE77 partner repository with the dataset identifier PXD021821. Processed sequencing data, as well as raw and processed metabolic and proteomics data are available at the dedicated online portal at https://artyomovlab.wustl.edu/aging/. The following publicly available datasets have been used in this study: GSE56045 MESA transcriptomic dataset—expression table and annotation are provided; GSE56046 MESA DNA methylation dataset—table of M values and annotation are provided; EGAD00001002523 blueprint datase—bigwig files with methylation signal were downloaded—experiment IDs are EGAX00001097775, EGAX00001097772, EGAX00001086967, EGAX00001086968, EGAX00001086970 and EGAX00001097774; GSE31263 WGBS dataset—BED files with methylation signal were used; ENCODE ChIP–seq samples were used (GSM1102782, GSM1102785, GSM1102788, GSM1102793 and GSM1102797); ENCODE ChromHMM annotation ENCSR907LCD was used (https://www.encodeproject.org/annotations/ENCSR907LCD/); ReMap 2018 database was downloaded for hg19 (http://pedagogix-tagc.univ-mrs.fr/remap/download/remap2018/hg19/MACS/remap2018_TF_archive_nr_macs2_hg19_v1_2.tar.gz); for a list of datasets used in Fig. 6 and Extended Data Fig. 9, see ‘Integration with public data’ in Methods; Neal lab analysis of UK Biobank was downloaded from http://www.nealelab.is/blog/2017/7/19/rapid-gwas-of-thousands-of-phenotypes-for-337000-samples-in-the-uk-biobank.

Code availability

The ULI-ChIP–seq data processing pipeline is available on GitHub: https://github.com/JetBrains-Research/washu. Custom tools (SPAN and JBR genome browser), models and tutorials are available online at https://artyomovlab.wustl.edu/aging/tools.html.

References

  1. Marengoni, A. et al. Aging with multimorbidity: a systematic review of the literature. Ageing Res. Rev. 10, 430–439 (2011).

    PubMed  Google Scholar 

  2. Bektas, A., Schurman, S. H., Sen, R. & Ferrucci, L. Aging, inflammation and the environment. Exp. Gerontol. 105, 10–18 (2018).

    CAS  PubMed  Google Scholar 

  3. Cesari, M. et al. Antioxidants and physical performance in elderly persons: the Invecchiare in Chianti (InCHIANTI) study. Am. J. Clin. Nutr. 79, 289–294 (2004).

    CAS  PubMed  Google Scholar 

  4. Reynolds, L. M. et al. Age-related variations in the methylome associated with gene expression in human monocytes and T cells. Nat. Commun. 5, 5366 (2014).

    PubMed  Google Scholar 

  5. Peters, M. J. et al. The transcriptional landscape of age in human peripheral blood. Nat. Commun. 6, 8570 (2015).

    CAS  PubMed  Google Scholar 

  6. Carr, E. J. et al. The cellular composition of the human immune system is shaped by age and cohabitation. Nat. Immunol. 17, 461–468 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  7. Patin, E. et al. Natural variation in the parameters of innate immune cells is preferentially driven by genetic factors. Nat. Immunol. 19, 302–314 (2018).

    CAS  PubMed  Google Scholar 

  8. Beerman, I. et al. Proliferation-dependent alterations of the DNA methylation landscape underlie hematopoietic stem cell aging. Cell Stem Cell 12, 413–425 (2013).

    CAS  PubMed  Google Scholar 

  9. Benayoun, B. A., Pollina, E. A. & Brunet, A. Epigenetic regulation of ageing: linking environmental inputs to genomic stability. Nat. Rev. Mol. Cell Biol. 16, 593–610 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  Google Scholar 

  11. Johnson, A. A. et al. The role of DNA methylation in aging, rejuvenation, and age-related disease. Rejuvenation Res. 15, 483–494 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  12. Zhang, W., Qu, J., Liu, G. H. & Belmonte, J. C. I. The ageing epigenome and its rejuvenation. Nat. Rev. Mol. Cell Biol. 21, 137–150 (2020).

    CAS  PubMed  Google Scholar 

  13. Brind’Amour, J. et al. An ultra-low-input native ChIP–seq protocol for genome-wide profiling of rare cell populations. Nat. Commun. 6, 6033 (2015).

    PubMed  Google Scholar 

  14. Brodin, P. et al. Variation in the human immune system is largely driven by non-heritable influences. Cell 160, 37–47 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  15. Trabado, S. et al. The human plasma-metabolome: reference values in 800 French healthy volunteers; impact of cholesterol, gender and age. PLoS ONE 12, e0173615 (2017).

    PubMed  PubMed Central  Google Scholar 

  16. Rodriguez-Rodero, S. et al. Aging genetics and aging. Aging Dis. 2, 186–195 (2011).

    PubMed  PubMed Central  Google Scholar 

  17. Mitchell, S. J., Scheibye-Knudsen, M., Longo, D. L. & de Cabo, R. Animal models of aging research: implications for human aging and age-related diseases. Annu. Rev. Anim. Biosci. 3, 283–303 (2015).

    CAS  PubMed  Google Scholar 

  18. Kim, H. O., Kim, H. S., Youn, J. C., Shin, E. C. & Park, S. Serum cytokine profiles in healthy young and elderly population assessed using multiplexed bead-based immunoassays. J. Transl. Med. 9, 113 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  19. Clark, J. A. & Peterson, T. C. Cytokine production and aging: overproduction of IL-8 in elderly males in response to lipopolysaccharide. Mech. Ageing Dev. 77, 127–139 (1994).

    CAS  PubMed  Google Scholar 

  20. Wolf, J. et al. The effect of chronological age on the inflammatory response of human fibroblasts. Exp. Gerontol. 47, 749–753 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  21. Franceschi, C., Garagnani, P., Parini, P., Giuliani, C. & Santoro, A. Inflammaging: a new immune-metabolic viewpoint for age-related diseases. Nat. Rev. Endocrinol. 14, 576–590 (2018).

    CAS  PubMed  Google Scholar 

  22. Mahlknecht, U. & Kaiser, S. Age-related changes in peripheral blood counts in humans. Exp. Ther. Med. 1, 1019–1025 (2010).

    PubMed  PubMed Central  Google Scholar 

  23. Geiger, H., de Haan, G. & Florian, M. C. The ageing haematopoietic stem cell compartment. Nat. Rev. Immunol. 13, 376–389 (2013).

    CAS  PubMed  Google Scholar 

  24. Conte, M. et al. Human aging and longevity are characterized by high levels of mitokines. J. Gerontol. A 74, 600–607 (2018).

    Google Scholar 

  25. Tanaka, T. et al. Plasma proteomic signature of age in healthy humans. Aging Cell 17, e12799 (2018).

    PubMed  PubMed Central  Google Scholar 

  26. Labrie, F., Belanger, A., Cusan, L., Gomez, J. L. & Candas, B. Marked decline in serum concentrations of adrenal C19 sex steroid precursors and conjugated androgen metabolites during aging. J. Clin. Endocrinol. Metab. 82, 2396–2402 (1997).

    CAS  PubMed  Google Scholar 

  27. Liu, Y. et al. Methylomics of gene expression in human monocytes. Hum. Mol. Genet. 22, 5065–5074 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  28. Reynolds, L. M. et al. Transcriptomic profiles of aging in purified human immune cells. BMC Genomics 16, 333 (2015).

    PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  30. Florath, I., Butterbach, K., Muller, H., Bewerunge-Hudler, M. & Brenner, H. Cross-sectional and longitudinal changes in DNA methylation with age: an epigenome-wide analysis revealing over 60 novel age-associated CpG sites. Hum. Mol. Genet. 23, 1186–1201 (2014).

    CAS  PubMed  Google Scholar 

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

    CAS  PubMed  Google Scholar 

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

    PubMed  PubMed Central  Google Scholar 

  33. Horvath, S. et al. Aging effects on DNA methylation modules in human brain and blood tissue. Genome Biol. 13, R97 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  34. Weidner, C. I. et al. Aging of blood can be tracked by DNA methylation changes at just three CpG sites. Genome Biol. 15, R24 (2014).

    PubMed  PubMed Central  Google Scholar 

  35. Garrett-Bakelman, F. E. et al. Enhanced reduced representation bisulfite sequencing for assessment of DNA methylation at base pair resolution. J. Vis. Exp. 96, e52246 (2015).

    Google Scholar 

  36. Heyn, H. et al. Distinct DNA methylomes of newborns and centenarians. Proc. Natl Acad. Sci. USA 109, 10522–10527 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  37. Wilson, V. L., Smith, R. A., Ma, S. & Cutler, R. G. Genomic 5-methyldeoxycytidine decreases with age. J. Biol. Chem. 262, 9948–9951 (1987).

    CAS  PubMed  Google Scholar 

  38. Cheung, P. et al. Single-cell chromatin modification profiling reveals increased epigenetic variations with aging. Cell 173, 1385–1397.e14 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  39. Slieker, R. C. et al. Age-related accrual of methylomic variability is linked to fundamental ageing mechanisms. Genome Biol. 17, 191 (2016).

    PubMed  PubMed Central  Google Scholar 

  40. Tserel, L. et al. Age-related profiling of DNA methylation in CD8+ T cells reveals changes in immune response and transcriptional regulator genes. Sci. Rep. 5, 13107 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  41. Song, Q. et al. A reference methylome database and analysis pipeline to facilitate integrative and comparative epigenomics. PLoS ONE 8, e81148 (2013).

    PubMed  PubMed Central  Google Scholar 

  42. Adams, D. et al. BLUEPRINT to decode the epigenetic signature written in blood. Nat. Biotechnol. 30, 224–226 (2012).

    CAS  PubMed  Google Scholar 

  43. Stunnenberg, H. G., International Human Epigenome Consortium & Hirst, M. The International Human Epigenome Consortium: a blueprint for scientific collaboration and discovery. Cell 167, 1145–1149 (2016).

    CAS  PubMed  Google Scholar 

  44. Hocking, T. D. et al. Optimizing ChIP–seq peak detectors using visual labels and supervised machine learning. Bioinformatics 33, 491–499 (2017).

    CAS  PubMed  Google Scholar 

  45. Ernst, J. & Kellis, M. Chromatin-state discovery and genome annotation with ChromHMM. Nat. Protoc. 12, 2478–2492 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  46. Cheneby, J., Gheorghe, M., Artufel, M., Mathelier, A. & Ballester, B. ReMap 2018: an updated atlas of regulatory regions from an integrative analysis of DNA-binding ChIP–seq experiments. Nucleic Acids Res. 46, D267–D275 (2018).

    CAS  PubMed  Google Scholar 

  47. Griffon, A. et al. Integrative analysis of public ChIP–seq experiments reveals a complex multi-cell regulatory landscape. Nucleic Acids Res. 43, e27 (2015).

    PubMed  Google Scholar 

  48. Fernandez, A. F. et al. H3K4me1 marks DNA regions hypomethylated during aging in human stem and differentiated cells. Genome Res. 25, 27–40 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  49. Gasparoni, G. et al. DNA methylation analysis on purified neurons and glia dissects age and Alzheimer’s disease-specific changes in the human cortex. Epigenetics Chromatin 11, 41 (2018).

    PubMed  PubMed Central  Google Scholar 

  50. Gross, A. M. et al. Methylome-wide analysis of chronic HIV infection reveals five-year increase in biological age and epigenetic targeting of HLA. Mol. Cell 62, 157–168 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  51. Wan, M. et al. Identification of smoking-associated differentially methylated regions using reduced representation bisulfite sequencing and cell type-specific enhancer activation and gene expression. Environ. Health Perspect. 126, 047015 (2018).

    PubMed  PubMed Central  Google Scholar 

  52. Day, S. E. et al. Next-generation sequencing methylation profiling of subjects with obesity identifies novel gene changes. Clin. Epigenetics 8, 77 (2016).

    PubMed  PubMed Central  Google Scholar 

  53. Marquez, E. J. et al. Sexual-dimorphism in human immune system aging. Nat. Commun. 11, 751 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  54. Vire, E. et al. The Polycomb group protein EZH2 directly controls DNA methylation. Nature 439, 871–874 (2006).

    CAS  PubMed  Google Scholar 

  55. Mozhui, K. & Pandey, A. K. Conserved effect of aging on DNA methylation and association with EZH2 Polycomb protein in mice and humans. Mech. Ageing Dev. 162, 27–37 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  56. Zhao, M. T. et al. Cell type-specific chromatin signatures underline regulatory DNA elements in human induced pluripotent stem cells and somatic cells. Circ. Res. 121, 1237–1250 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  57. Baker, D. J. et al. Opposing roles for p16Ink4a and p19Arf in senescence and ageing caused by BubR1 insufficiency. Nat. Cell Biol. 10, 825–836 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  58. Gold, L. et al. Aptamer-based multiplexed proteomic technology for biomarker discovery. PLoS ONE 5, e15004 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  59. Ritchie, M. E. et al. limma powers differential expression analyses for RNA-sequencing and microarray studies. Nucleic Acids Res. 43, e47 (2015).

    PubMed  PubMed Central  Google Scholar 

  60. Yang, J. et al. Synchronized age-related gene expression changes across multiple tissues in human and the link to complex diseases. Sci. Rep. 5, 15145 (2015).

    PubMed  PubMed Central  Google Scholar 

  61. Sergushichev, A. A. An algorithm for fast preranked gene set enrichment analysis using cumulative statistic calculation. Preprint at bioRxiv https://doi.org/10.1101/060012 (2016).

  62. Jha, A. K. et al. Network integration of parallel metabolic and transcriptional data reveals metabolic modules that regulate macrophage polarization. Immunity 42, 419–430 (2015).

    CAS  PubMed  Google Scholar 

  63. Derr, A. et al. End sequence analysis toolkit (ESAT) expands the extractable information from single-cell RNA-seq data. Genome Res. 26, 1397–1410 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  64. Storey, J. D. & Tibshirani, R. Statistical significance for genomewide studies. Proc. Natl Acad. Sci. USA 100, 9440–9445 (2003).

    CAS  PubMed  PubMed Central  Google Scholar 

  65. Karolchik, D. et al. The UCSC Table Browser data retrieval tool. Nucleic Acids Res. 32, D493–D496 (2004).

    CAS  PubMed  PubMed Central  Google Scholar 

  66. Legrand, C. et al. Statistically robust methylation calling for whole-transcriptome bisulfite sequencing reveals distinct methylation patterns for mouse RNAs. Genome Res. 27, 1589–1596 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  67. Yu, G., Wang, L. G. & He, Q. Y. ChIPseeker: an R/Bioconductor package for ChIP peak annotation, comparison and visualization. Bioinformatics 31, 2382–2383 (2015).

    CAS  PubMed  Google Scholar 

  68. Quinlan, A. R. & Hall, I. M. BEDTools: a flexible suite of utilities for comparing genomic features. Bioinformatics 26, 841–842 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  69. Quinlan, A. R. BEDTools: the Swiss-army tool for genome feature analysis. Curr. Protoc. Bioinformatics 47, 1.12.1–1.12.34 (2014).

    Google Scholar 

  70. Thorvaldsdottir, H., Robinson, J. T. & Mesirov, J. P. Integrative Genomics Viewer (IGV): high-performance genomics data visualization and exploration. Brief. Bioinform. 14, 178–192 (2013).

    CAS  PubMed  Google Scholar 

  71. Robinson, J. T. et al. Integrative genomics viewer. Nat. Biotechnol. 29, 24–26 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  72. Du, P., Kibbe, W. A. & Lin, S. M. lumi: a pipeline for processing Illumina microarray. Bioinformatics 24, 1547–1548 (2008).

    CAS  PubMed  Google Scholar 

  73. Consortium, E. P. An integrated encyclopedia of DNA elements in the human genome. Nature 489, 57–74 (2012).

    Google Scholar 

  74. Steinhauser, S., Kurzawa, N., Eils, R. & Herrmann, C. A comprehensive comparison of tools for differential ChIP–seq analysis. Brief. Bioinform. 17, 953–966 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  75. Nicodemus-Johnson, J. et al. DNA methylation in lung cells is associated with asthma endotypes and genetic risk. JCI Insight 1, e90151 (2016).

    PubMed  PubMed Central  Google Scholar 

  76. Lunnon, K. et al. Methylomic profiling implicates cortical deregulation of ANK1 in Alzheimer’s disease. Nat. Neurosci. 17, 1164–1170 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  77. Perez-Riverol, Y. et al. The PRIDE database and related tools and resources in 2019: improving support for quantification data. Nucleic Acids Res. 47, D442–D450 (2019).

    CAS  PubMed  Google Scholar 

Download references

Acknowledgements

The study was supported by funding from the Aging Biology Foundation to the Artyomov laboratory. The Bagaitkar lab is partially supported by GM125504 and DE28296. The Dixit lab is supported in part by NIH grants P01AG051459, AI105097, AG051459 and AR070811, the Glenn Foundation on Aging Research and Cure Alzheimer’s Fund. This publication is solely the responsibility of the authors and does not necessarily represent the official view of the National Centre for Research Resources (NCRR) or the National Institutes of Health (NIH). We thank the Genome Technology Access Centre in the Department of Genetics at Washington University School of Medicine for help with genomic analysis. The centre is partially supported by NCI Cancer Centre Support grant number P30 CA91842 to the Siteman Cancer Centre and by ICTS/CTSA grant number UL1TR000448 from the NCRR, a component of the NIH, and the NIH Roadmap for Medical Research. We also thank the Epigenomic Core of Weill Cornell Medicine for the initial analysis of the methylation data (eRRBS and raw data pre-processing). We acknowledge the ENCODE consortium and the ENCODE production laboratories that generated the datasets used in the manuscript. We thank I. Miralda for the Fig. 1 schematic.

Author information

Authors and Affiliations

Authors

Contributions

I.S., J.B. and O.S. contributed equally. I.S., J.B., O.S., S.D. and M.N.A. designed the project. J.B., D.A.M., E.L., S.P., L.A., A.S. and A.P. performed wet laboratory experiments. I.S., J.B, O.S., E.L., S.P., D.A.M., E.W., P.C., G.D., M.A., K.Z., S.S., C.C., M.B., L.A., A.S., A.P., A.D. E.K., P.T., R.C. V.D.D., M.J., S.D., S.A.S., M.J.D., E.M.O. and M.N.A. participated in data collection and data analysis. I.S., J.B., O.S., G.D, M.A., E.K., P.T. and R.C. made the figures and tables. Writing and editing were done by I.S., J.B., O.S. and M.N.A.

Corresponding author

Correspondence to Maxim N. Artyomov.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Peer review information Nature Aging thanks the anonymous reviewer(s) 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

Extended Data Fig. 1 Cell counts and cytokine profiles.

a, Blood cytokine levels measured by bioplex assay and b, blood differentials obtained using Hemavet in young (n = 20) and old (n = 20) cohorts. Normal ranges for humans are shown below the boxplots. P-values for all comparisons were calculated using two-sided Mann-Whitney U test. In both panels, the lower and upper hinges of all boxplots represent the 25th and 75th percentiles. Horizontal bars show median value. Whiskers extend to the values that are no further than 1.5*IQR from either upper or lower hinge. IQR stands for inter-quartile range, which is the difference between the 75th and 25th percentiles.

Extended Data Fig. 2 Metabolomic profile of plasma.

a, PCA of standardized levels of metabolites in plasma. Each dot represents a donor. b, Dendrogram produced by unsupervised hierarchical clustering of metabolic data. Clustering using average algorithm and Euclidian distance as the distance metric. c, List of significantly different metabolites. d, Selected differentially regulated metabolites from sex steroids synthesis pathway in young (n = 20) and old (n = 20) cohorts. Statistical analyses by two-sided Mann-Whitney U test with Benjamini–Hochberg correction for multiple testing. e, Schema of sex steroids synthesis. f, Pathway analysis of metabolic data. Each boxplot summarizes log2FC of all members of the corresponding pathway. Pathways with mean log2FC significantly different from zero are highlighted (two-sided Mann-Whitney U test and Benjamini-Hochberg correction for multiple testing). Dots represent outliers. N for each pathway is shown in brackets. g, PCA as in Extended Data 2a. Z-scores were calculated for all metabolites. For each sample, color of the dot represents averaged z-scores of all metabolites belonging to the pathway. In panels (D) and (F), the lower and upper hinges of all boxplots represent the 25th and 75th percentiles. Horizontal bars show median value. Whiskers extend to the values that are no further than 1.5*IQR from either upper or lower hinge. IQR stands for inter-quartile range, which is the difference between the 75th and 25th percentiles.

Extended Data Fig. 3 Proteomic profile of plasma (Somascan).

a, PCA of standardized proteins levels in plasma (Somascan). Each dot represents a donor. b, Dendrogram produced by unsupervised hierarchical clustering of Somascan data. Clustering using average algorithm and Euclidian distance as the distance metric. c, Differential analysis results for plasma proteomic profile: volcano plot. Each dot represents a protein. P-values and logFC were calculated by two-sided test from Limma package and adjusted by Benjamini–Hochberg method. d, ELISA validation of selected proteins in young (n = 20) and old (n = 20) cohorts. Adjusted p-values for Somascan results (as in (C)) are shown in the top panel and ELISA validation of the same targets are shown below. ELISA p-values were calculated using two-sided Mann-Whitney U test. The lower and upper hinges of all boxplots represent the 25th and 75th percentiles. Horizontal bars show median value. Whiskers extend to the values that are no further than 1.5*IQR from either upper or lower hinge. IQR stands for inter-quartile range, which is the difference between the 75th and 25th percentiles. e, Heatmap representation of absolute values of Spearman’s correlation coefficients (rho) between plasma proteins (rows) and plasma metabolites (columns) calculated within old (left panel) and young (right panel) cohorts. Clustering of old cohort rows and columns was done using complete algorithm and Euclidian distance as a metric. Order of rows and columns in young cohort heatmap matches order established for the old cohort. f, Each point represents a donor. Smoothing was done by lm function separately in young and old groups, shaded error bands represent SE.

Extended Data Fig. 4 Monocyte transcriptome and proteome.

(a) Left panel: schematic representation of CD14+CD16 monocytes isolation using magnetic beads. Right panel: flow cytometry validation and estimation of purity. (b) Number of significant genes detected after downsampling MESA dataset (youngest and oldest 25%). Downsampling was repeated n = 50 times for each group size. The lower and upper hinges of all boxplots represent the 25th and 75th percentiles. Horizontal bars show median value. Whiskers extend to the values that are no further than 1.5*IQR from either upper or lower hinge. IQR stands for inter-quartile range, which is the difference between the 75th and 25th percentiles. (c) GSEA enrichment curves illustrate pathways that significantly change with age in MESA dataset. P-values are one-sided and corrected by Benjamini-Hochberg method. (d) Monocytes were differentiated into macrophages by one-week incubation with M-CSF. Both cell types were stimulated by LPS for 24 hours. (e) PCA of normalized expression levels estimated by RNA-seq for monocyte differentiation and activation experiment. (f) Each dot represents a monocyte protein significantly different between age groups. LogFC proteomics (x axis) as in Fig. 2f, LogFC transcriptomics as in Fig. 2c.

Extended Data Fig. 5 RRBS quality control and monocyte methylome.

a, Library depth for each sample. b, Hannum and Horvath methylation clocks for old (n = 20) and young (n = 20) groups. Methylation levels of CpGs that were used in the model but were not covered in our eRRBS data were imputed using mean methylation of [−100kb; +100 kb] region around the CpG. CpG methylation was set to zero if imputation was not possible. P-values were calculated using two-sided Mann-Whitney U test. The lower and upper hinges of all boxplots represent the 25th and 75th percentiles. Horizontal bars show median value. Whiskers extend to the values that are no further than 1.5*IQR from either upper or lower hinge. IQR stands for inter-quartile range, which is the difference between the 75th and 25th percentiles. c, Enrichment of DMRs in CpG islands. Histogram shows distribution of simulated intersection sizes (n = 100,000 random simulations). d, Comparison to Blueprint dataset. Each dot represents one DMR detected in our dataset. X axis – difference between old and young cohorts in our dataset, Y axis – difference between old donors and cord blood from Blueprint. e, Plot as in right panel of Fig. 3g for a newborn vs centenarian WGBS dataset (GSE31263). f, PCA on MESA data as in Fig. 3i using all cytosines profiled by DNA methylation array. g, Number of methylated (methylation level > 0) cytosines in CpG, CHG and CHH context shared by one to 40 samples. h, PCA of CpGs methylation levels from old and young groups. Each dot represents a sample. i, Dendrogram produced by unsupervised hierarchical clustering of the samples. Each sample described as a vector of CpG methylation levels. Clustering using Ward algorithm and Manhattan distance as the distance metric. Outliers are labelled. j, Distribution of CpG methylation levels. For each segment fractions of CpGs with corresponding methylation level in each sample are shown by dots. Bar shows average fraction of CpG across all samples. Outlying samples are labelled. k, PCA of CpGs methylation levels. Outliers (OD11, OD17, YD9) were excluded.

Extended Data Fig. 6 ULI-ChIP-seq processing and quality control.

a, Snapshot of the H3K4me1 tracks across all donors shows distinct signal for all samples with high variability in the signal-to-noise ratio. b, Number of peaks for each mark in each donor yielded by classical peak calling tools. c, Schematic representation of overlap metric used in panels (E) and (H). d, Number of peaks yielded by SPAN, MACS2 and SICER. e, Overlap between all pairs of samples for peaks generated by SPAN, MACS2 and SICER. SICER was used for wide modifications only. N for each bar is equal to a number of possible pairs between all samples that passed QC. f, Summary for panel (E). Mean and standard deviation (SD) of overlaps between samples are shown. g, Overlap characteristics as in (F) for SPAN runs with various annotation sizes. h, Directional overlap of SPAN peaks between all samples and all histone modifications. i, Two-way overlap with ENCODE CD14+ monocytes data for different peak calling approaches. In panels (B), (D), and (I) N for each bar is equal to a number of ChIP-seq samples that passed QC for each modification. See Fig. 4b for exact numbers. Error bars in all panels represent SD.

Extended Data Fig. 7 SPAN test set errors.

Test set errors for golden standard tools (MACS2, SICER) as well as SPAN trained with various numbers of labels. Each dot represents a sample, n = 40 for H3K4me3, H3K27me3, H3K27ac, n = 32 for H3K4me1, n = 39 for H3K36me3. In all panels, the lower and upper hinges of all boxplots represent the 25th and 75th percentiles. Horizontal bars show median value. Whiskers extend to the values that are no further than 1.5*IQR from either upper or lower hinge. IQR stands for inter-quartile range, which is the difference between the 75th and 25th percentiles.

Extended Data Fig. 8 Chromatin context of DMRs.

a, Enrichment of DMRs in chromatin state segments from ENCODE ChromHMM partition. Upper: for each chromatin state dark grey bar represents number of DMRs intersecting at least one segment of the state. Light grey bar shows expected number of intersections estimated by n = 100,000 random simulations. Error bars show SD, their centers represent expected intersection. Results shown for all DMRs, hypermethylated DMRs only and hypomethylated DMRs only. Chromatin states that are significantly over- or under-represented among DMRs are marked by asterix (*). Bottom: heatmap shows general statistics for each chromatin state. Values are normalized within each row. b, PCA of standardized methylation levels in old and young groups for three chromatin states: bivalent Tss (14 TssBiv), bivalent enhancers (15 EnhBiv) and Polycomb-repressed regions (16 ReprPC). Each dot represents a sample. c, Intensity of H3K4me1, H3K27me3, H3K4me3, H3K27ac, and H3K36me3 signals was calculated for each DMR via Diffbind package, normalized with respect to the DMR length and averaged across the cohorts. Normalized signals were compared between hypo- (n = 423) and hypermethylated (n = 737) DMRs using two-sided Mann-Whitney U test. Dots represent outliers. The lower and upper hinges of all boxplots represent the 25th and 75th percentiles. Horizontal bars show median value. Whiskers extend to the values that are no further than 1.5*IQR from either upper or lower hinge. IQR stands for inter-quartile range, which is the difference between the 75th and 25th percentiles. d, Volcano plot as in Figs. 4i and 4k, colored in accordance with intersection with H3K4me3, H3K27ac and H3K36me3. e, Density plot protein coding gene expression with transcription factors of interest highlighted.

Extended Data Fig. 9 Up and down DMRs in different physiological conditions.

a, Comparison of age- and sex-adjusted DMR mean methylation between Alzheimer’s patients (n = 15 for glia and neurons, n = 48 for whole blood) and healthy controls (n = 14 for glia and neurons, n = 9 for whole blood). Each dot represents one donor. P-values calculated using two-sided Mann-Whitney U test. b, Plot as in (A) comparing data from lean (n = 9) and obese (n = 8) donors. c, Plot as in Fig. 6g. Mean methylation of smoking DMRs in our cohort: young lean (n = 11), young overweight (n = 8), old lean (n = 8), and old overweight (n = 10). P-values were calculated using two-sided Mann-Whitney U test. In all panels, the lower and upper hinges of all boxplots represent the 25th and 75th percentiles. Horizontal bars show median value. Whiskers extend to the values that are no further than 1.5*IQR from either upper or lower hinge. IQR stands for inter-quartile range, which is the difference between the 75th and 25th percentiles.

Supplementary information

Supplementary Information

Supplementary Methods and Supplementary Figs. 1 and 2.

Reporting Summary

Supplementary Table 1

Basic donor information and blood differential (cell counts, Hb and HCT levels).

Supplementary Table 2

Cytokine bioplex assay data for all donors (IFN-γ, IL-10, IL-12p70, IL-13, IL-1β, IL-2, IL-4, IL-6, IL-8, TNF-α).

Supplementary Table 3

(A) Scaled intensities of the metabolites in the plasma for all donors (734 metabolites). (B) Differential analysis results. Significant differences in metabolites between cohorts were determined using two-sided Mann–Whitney U-test. P values were adjusted for multiple testing using the Benjamini–Hochberg method.

Supplementary Table 4

SomaScan proteomics array data for all donors: (A) scaled data, (B) differential comparison statistics. For differential analysis, functions lmFit and eBayes from the Limma package were used (two-sided). P values were adjusted for multiple testing using the Benjamini–Hochberg method.

Supplementary Table 5

ELISA validation of the most differentially present plasma proteins (sCD86, GDF-15, sclerostin, OMD, Notch1).

Supplementary Table 6

Counts for monocyte RNA-seq data. (A) Raw counts, (B) DESeq2 normalized counts and (C) log2-quantile-normalized counts.

Supplementary Table 7

Monocyte RNA-seq data. DESeq2 differential analysis results between old and young cohorts (two-sided Wald test, correction for multiple testing using the Benjamini–Hochberg method).

Supplementary Table 8

Transcriptomic data for MESA cohort. Limma differential analysis results (gene ~ age + chip + race–gender–site design, two-sided test, correction for multiple testing using the Benjamini–Hochberg method).

Supplementary Table 9

Monocyte proteomics data generated and analysed by Biognosys. (A) Sample table, (B) spectral library protein inventory, (C) spectral library peptide inventory, (D) differential analysis results, (E) protein intensities and (F) peptide intensities. In brief, for each protein, the fold change of each peptide ion variant was estimated as average abundance of peptide ion variant across biological replicates in the older group/average abundance of peptide ion variant across biological replicates in the young group. The values then were log-transformed and fold changes of all peptides belonging to the same proteins were compared to zero using two-sided paired t-test. Multiple testing correction was performed as described in Storey et al.63.

Supplementary Table 10

RNA-seq data for monocyte differentiation and stimulation experiment (Extended Data 4D). (A) Raw counts, (B) DESeq2-normalized counts and (C) log2-quantile-normalized counts.

Supplementary Table 11

Basic QC metrics of RRBS libraries (n covered (≥ 10 reads) CpG, mean CpG coverage, percent mapped reads, conversion rate and library depth).

Supplementary Table 12

Differentially methylated regions as obtained from RRBS data: (A) unfiltered DMRs and (B) Confident DMRs filtered based on ncyto ≥ 3 and abs(avdiff) ≥ 0.025.

Supplementary Table 13

Basic characteristics and QC of the ULI-ChIP–seq libraries: (A) FastqC output, (B) QC of bam files based on ENCODE standards, (C) alignment statistics and (D) QC results reported by phantompeaks.

Supplementary Table 14

Transcription factors binding sites overrepresentation analysis: (A) results for up DMRs corrected for enrichment in CpG islands and (B) results for down DMRs corrected for enrichment in H3K4me1-marked regions. See Methods for the details of the one-sided enrichment procedure. Correction for multiple testing was done using the Benjamini–Hochberg method.

Supplementary Table 15

Positive and negative primers used for the ChIP qPCR quality control.

Supplementary Table 16

Peak calling summary for all samples (MACS2, SICER and SPAN).

Supplementary Table 17

Labels used for the semi-supervised peak calling: (A) overall label sets statistics. Specific labels for (B) H3K27ac, (C) H3K27me3, (D) H3K36me3, (E) H3K4me1 and (F) H3K4me3.

Supplementary Table 18

Comparisons of errors produced by SICER, MACS2 and SPAN on the datasets annotated by McGill team from Hocking et al.44.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Shchukina, I., Bagaitkar, J., Shpynov, O. et al. Enhanced epigenetic profiling of classical human monocytes reveals a specific signature of healthy aging in the DNA methylome. Nat Aging 1, 124–141 (2021). https://doi.org/10.1038/s43587-020-00002-6

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s43587-020-00002-6

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