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.

  • Article
  • Published:

Multi-omic rejuvenation and lifespan extension on exposure to youthful circulation

Abstract

Heterochronic parabiosis (HPB) is known for its functional rejuvenation effects across several mouse tissues. However, its impact on biological age and long-term health is unknown. Here we performed extended (3-month) HPB, followed by a 2-month detachment period of anastomosed pairs. Old detached mice exhibited improved physiological parameters and lived longer than control isochronic mice. HPB drastically reduced the epigenetic age of blood and liver based on several clock models using two independent platforms. Remarkably, this rejuvenation effect persisted even after 2 months of detachment. Transcriptomic and epigenomic profiles of anastomosed mice showed an intermediate phenotype between old and young, suggesting a global multi-omic rejuvenation effect. In addition, old HPB mice showed gene expression changes opposite to aging but akin to several lifespan-extending interventions. Altogether, we reveal that long-term HPB results in lasting epigenetic and transcriptome remodeling, culminating in the extension of lifespan and healthspan.

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: Prolonged parabiosis followed by detachment leads to extended lifespan and healthspan.
Fig. 2: Persistent epigenetic age reversal in blood and liver on HPB assessed by RRBS-based aging clocks.
Fig. 3: Sustained epigenetic age reversal in liver on HPB assessed by microarray-based aging clocks.
Fig. 4: Transcriptomic analyses of HPB reveal unique pathway enrichment and differential expression patterns.
Fig. 5: Dimensionality reduction highlights rejuvenated molecular profiles after HPB.
Fig. 6: The transcriptomic signatures of HPB align with longevity interventions and oppose aging.
Fig. 7: Gene expression analyses hint at putative rejuvenation mechanisms of long-term HPB.

Similar content being viewed by others

Data availability

The sequencing data obtained in this study are deposited with the Gene Expression Omnibus under accession no. GSE224447. The data used to generate the graphs can be found in the accompanying source data files. Source data for Figs. 17 and Extended Data Figs. 110 are provided with the paper.

References

  1. Brett, J. O. & Rando, T. A. Alive and well? Exploring disease by studying lifespan. Curr. Opin. Genet. Dev. 26, 33–40 (2014).

    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. López-Otín C., Blasco, M. A., Partridge, L., Serrano, M. & Kroemer, G. Hallmarks of aging: an expanding universe. Cell 186, 243–278 (2023).

    Article  PubMed  Google Scholar 

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

    Article  PubMed  PubMed Central  Google Scholar 

  5. 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 

  6. 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 

  7. 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 

  8. Fahy, G. M. et al. Reversal of epigenetic aging and immunosenescent trends in humans. Aging Cell 18, e13028 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Horvath, S. et al. Reversing age: dual species measurement of epigenetic age with a single clock. Preprint at bioRxiv https://doi.org/10.1101/2020.05.07.082917 (2020).

  10. 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 

  11. Rando, T. A. & Chang, H. Y. Aging, rejuvenation, and epigenetic reprogramming: resetting the aging clock. Cell 148, 46–57 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. 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 

  13. 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 

  14. Trapp, A., Kerepesi, C. & Gladyshev, V. N. Profiling epigenetic age in single cells. Nat. Aging 1, 1189–1201 (2021).

    Article  PubMed  PubMed Central  Google Scholar 

  15. Lunsford, W. R., McCay, C. C., Lupien, P. J., Pope, F. E. & Sperling, G. Parabiosis as a method for studying factors which affect aging in rats. Gerontologia 7, 1–8 (1963).

    Article  CAS  PubMed  Google Scholar 

  16. McCay, C. M., Pope, F. & Lunsford, W. Experimental prolongation of the life span. Bull. N. Y. Acad. Med. 32, 91–101 (1956).

    CAS  PubMed  PubMed Central  Google Scholar 

  17. Pope, F., Lunsford, W. & McCay, C. M. Experimental prolongation of the life span. J. Chronic Dis. 4, 153–158 (1956).

    Article  CAS  PubMed  Google Scholar 

  18. Baht, G. S. et al. Exposure to a youthful circulation rejuvenates bone repair through modulation of β-catenin. Nat. Commun. 6, 7131 (2015).

    Article  CAS  PubMed  Google Scholar 

  19. Conboy, I. M. et al. Rejuvenation of aged progenitor cells by exposure to a young systemic environment. Nature 433, 760–764 (2005).

    Article  CAS  PubMed  Google Scholar 

  20. Loffredo, F. S. et al. Growth differentiation factor 11 is a circulating factor that reverses age-related cardiac hypertrophy. Cell 153, 828–839 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Ruckh, J. M. et al. Rejuvenation of regeneration in the aging central nervous system. Cell Stem Cell 10, 96–103 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Villeda, S. A. et al. Young blood reverses age-related impairments in cognitive function and synaptic plasticity in mice. Nat. Med. 20, 659–663 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Vi, L. et al. Macrophage cells secrete factors including LRP1 that orchestrate the rejuvenation of bone repair in mice. Nat. Commun. 9, 5191 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  24. Rebo, J. et al. A single heterochronic blood exchange reveals rapid inhibition of multiple tissues by old blood. Nat. Commun. 7, 13363 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Middeldorp, J. et al. Preclinical assessment of young blood plasma for Alzheimer disease. JAMA Neurol. 73, 1325–1333 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  26. Conboy, M. J., Conboy, I. M. & Rando, T. A. Heterochronic parabiosis: historical perspective and methodological considerations for studies of aging and longevity. Aging Cell 12, 525–530 (2013).

    Article  CAS  PubMed  Google Scholar 

  27. Wright, D. E., Wagers, A. J., Gulati, A. P., Johnson, F. L. & Weissman, I. L. Physiological migration of hematopoietic stem and progenitor cells. Science 294, 1933–1936 (2001).

    Article  CAS  PubMed  Google Scholar 

  28. Donskoy, E. & Goldschneider, I. Thymocytopoiesis is maintained by blood-borne precursors throughout postnatal life. A study in parabiotic mice. J. Immunol. 148, 1604–1612 (1992).

    Article  CAS  PubMed  Google Scholar 

  29. Ho, T. T. et al. Aged hematopoietic stem cells are refractory to bloodborne systemic rejuvenation interventions. J. Exp. Med. 218, e20210223 (2021).

  30. 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 

  31. 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 

  32. Lu, A. T. et al. Universal DNA methylation age across mammalian tissues. Nat. Aging https://doi.org/10.1038/s43587-023-00462-6 (2023).

  33. Arneson, A. et al. A mammalian methylation array for profiling methylation levels at conserved sequences. Nat. Commun. 13, 783 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Fiorito, G. et al. DNA methylation-based biomarkers of aging were slowed down in a two-year diet and physical activity intervention trial: the DAMA study. Aging Cell 20, e13439 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Durieux, J., Wolff, S. & Dillin, A. The cell-non-autonomous nature of electron transport chain-mediated longevity. Cell 144, 79–91 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Loerch, P. M. et al. Evolution of the aging brain transcriptome and synaptic regulation. PLoS ONE 3, e3329 (2008).

    Article  PubMed  PubMed Central  Google Scholar 

  37. Subramanian, A. et al. Gene set enrichment analysis: a knowledge-based approach for interpreting genome-wide expression profiles. Proc. Natl Acad. Sci. USA 102, 15545–15550 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Liberzon, A. et al. The molecular signatures database (MSigDB) hallmark gene set collection. Cell Syst. 1, 417–425 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Lesnefsky, E. J. & Hoppel, C. L. Oxidative phosphorylation and aging. Ageing Res. Rev. 5, 402–433 (2006).

    Article  CAS  PubMed  Google Scholar 

  40. Bandres, E. et al. The increase of IFN-gamma production through aging correlates with the expanded CD8+highCD28CD57+ subpopulation. Clin. Immunol. 96, 230–235 (2000).

    Article  CAS  PubMed  Google Scholar 

  41. Leonardi, G. C., Accardi, G., Monastero, R., Nicoletti, F. & Libra, M. Ageing: from inflammation to cancer. Immun. Ageing 15, 1 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  42. Lai, K. S. P. et al. Peripheral inflammatory markers in Alzheimer’s disease: a systematic review and meta-analysis of 175 studies. J. Neurol. Neurosurg. Psychiatry 88, 876–882 (2017).

    Article  PubMed  Google Scholar 

  43. Amdur, R. L. et al. Inflammation and progression of CKD: the CRIC study. Clin. J. Am. Soc. Nephrol. 11, 1546–1556 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Tyshkovskiy, A. et al. Identification and application of gene expression signatures associated with lifespan extension. Cell Metab. 30, 573–593 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Tyshkovskiy, A. et al. Distinct longevity mechanisms across and within species and their association with aging. Cell https://doi.org/10.1016/j.cell.2023.05.002 (2023).

  46. Gonzalez Herrera, K. N., Finley, L. W. & Haigis, M. C. The role of SIRT3 in regulating cancer cell metabolism. BMC Proc. 6, P18 (2012).

    Article  PubMed Central  Google Scholar 

  47. Benigni, A. et al. Sirt3 deficiency shortens life span and impairs cardiac mitochondrial function rescued by Opa1 gene transfer. Antioxid. Redox Signal. 31, 1255–1271 (2019).

    Article  CAS  PubMed  Google Scholar 

  48. Brown, K. et al. SIRT3 reverses aging-associated degeneration. Cell Rep. 3, 319–327 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Lang, C. A., Wu, W. K., Chen, T. & Mills, B. J. Blood glutathione: a biochemical index of life span enhancement in the diet restricted Lobund-Wistar rat. Prog. Clin. Biol. Res. 287, 241–246 (1989).

    CAS  PubMed  Google Scholar 

  50. Richie, J. P. Jr et al. Methionine restriction increases blood glutathione and longevity in F344 rats. FASEB J. 8, 1302–1307 (1994).

  51. Bernardes de Jesus, B. et al. Telomerase gene therapy in adult and old mice delays aging and increases longevity without increasing cancer. EMBO Mol. Med. 4, 691–704 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Okano, M., Bell, D. W., Haber, D. A. & Li, E. DNA methyltransferases Dnmt3a and Dnmt3b are essential for de novo methylation and mammalian development. Cell 99, 247–257 (1999).

    Article  CAS  PubMed  Google Scholar 

  53. Childs, B. G., Durik, M., Baker, D. J. & van Deursen, J. M. Cellular senescence in aging and age-related disease: from mechanisms to therapy. Nat. Med. 21, 1424–1435 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Conboy, I. M. & Rando, T. A. Heterochronic parabiosis for the study of the effects of aging on stem cells and their niches. Cell Cycle 11, 2260–2267 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Yankova, T., Dubiley, T., Shytikov, D. & Pishel, I. Three month heterochronic parabiosis has a deleterious effect on the lifespan of young animals, without a positive effect for old animals. Rejuvenation Res. 25, 191–199 (2022).

    Article  PubMed  Google Scholar 

  56. Castellano, J. M. et al. Human umbilical cord plasma proteins revitalize hippocampal function in aged mice. Nature 544, 488–492 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Salpeter, S. J. et al. Systemic regulation of the age-related decline of pancreatic β-cell replication. Diabetes 62, 2843–2848 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Villeda, S. A. et al. The ageing systemic milieu negatively regulates neurogenesis and cognitive function. Nature 477, 90–94 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  PubMed  PubMed Central  Google Scholar 

  60. Pálovics, R. et al. Molecular hallmarks of heterochronic parabiosis at single-cell resolution. Nature 603, 309–314 (2022).

  61. Conese, M., Carbone, A., Beccia, E. & Angiolillo, A. The fountain of youth: a tale of parabiosis, stem cells, and rejuvenation. Open Med. 12, 376–383 (2017).

    Article  CAS  Google Scholar 

  62. Elabd, C. et al. Oxytocin is an age-specific circulating hormone that is necessary for muscle maintenance and regeneration. Nat. Commun. 5, 4082 (2014).

    Article  CAS  PubMed  Google Scholar 

  63. Stölzel, F. et al. Dynamics of epigenetic age following hematopoietic stem cell transplantation. Haematologica 102, e321–e323 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  64. Mehdipour, M. et al. Rejuvenation of three germ layers tissues by exchanging old blood plasma with saline-albumin. Aging 12, 8790–8819 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Baht, G. S. et al. Meteorin-like facilitates skeletal muscle repair through a Stat3/IGF-1 mechanism. Nat. Metab. 2, 278–289 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. 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 

  67. Dobin, A. et al. STAR: ultrafast universal RNA-seq aligner. Bioinformatics 29, 15–21 (2013).

    Article  CAS  PubMed  Google Scholar 

  68. Liao, Y., Smyth, G. K. & Shi, W. featureCounts: an efficient general purpose program for assigning sequence reads to genomic features. Bioinformatics 30, 923–930 (2014).

    Article  CAS  PubMed  Google Scholar 

  69. Love, M. I., Huber, W. & Anders, S. Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2. Genome Biol. 15, 550 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  70. Robinson, M. D., McCarthy, D. J. & Smyth, G. K. edgeR: a Bioconductor package for differential expression analysis of digital gene expression data. Bioinformatics 26, 139–140 (2010).

    Article  CAS  PubMed  Google Scholar 

  71. Benjamini, Y. & Hochberg, Y. Controlling the false discovery rate: a practical and powerful approach to multiple testing. J. R. Stat. Soc. B 57, 289–300 (1995).

  72. Anders, S. & Huber, W. Differential expression analysis for sequence count data. Genome Biol. 11, R106 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  73. Huffman, K. M. et al. Exercise protects against cardiac and skeletal muscle dysfunction in a mouse model of inflammatory arthritis. J. Appl. Physiol. 130, 853–864 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  74. White, J. P. et al. The AMPK/p27Kip1 axis regulates autophagy/apoptosis decisions in aged skeletal muscle stem cells. Stem Cell Reports 11, 425–439 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

J.P.W. was supported by National Institutes of Health (NIH) grant nos. K01AG056664 and R21AG065943. V.N.G. was supported by NIH grant nos. R01AG067782, P01AG047200 and R01AG065403. D.E.L. was supported by NIH training grant no. T32HL007057. S.H. acknowledges support from the Milky Way Research Foundation and the Epigenetic Clock Development Foundation, and A. Tyshkovskiy and S.E.D. acknowledge support from the Interdisciplinary Scientific and Educational School of Moscow University ‘Molecular Technologies of the Living Systems and Synthetic Biology’. We thank the Duke Behavioral Core for support on this project. We also thank T. Fox for help with schematic figures. Figure icons were partially created with BioRender.com. The funders had no role in study design, data collection, data analysis, decision to publish, or preparation of the manuscript.

Author information

Authors and Affiliations

Authors

Contributions

B.Z., D.E.L., V.N.G. and J.P.W. conceived the project. D.E.L., G.S.B., A.B., L.K.M. and J.P.W. carried out the animal experiments. A. Trapp, D.E.L. and B.Z. designed the figures. A. Trapp, A. Tyshkovskiy and B.Z. performed the gene expression analyses. B.Z., A. Trapp and C.K. conducted the DNA methylation clock analyses based on RRBS. B.Z., A. Trapp, A.T.L. and S.H. conducted the analyses based on the microarrays. A.V.S. contributed to the enrichment analyses. S.E.D. contributed to data interpretation. V.N.G. and J.P.W. supervised the project. B.Z., D.E.L., A. Trapp, V.N.G. and J.P.W. wrote the paper with final approval from all coauthors.

Corresponding authors

Correspondence to Vadim N. Gladyshev or James P. White.

Ethics declarations

Competing interests

The authors declare no competing interests.

Peer review

Peer review information

Nature Aging thanks the anonymous reviewers for their contribution to the peer review of this work.

Additional information

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

Extended data

Extended Data Fig. 1 Timeline of molecular and physiological profiling.

a, Schematic timeline of the molecular (transcriptomic & epigenetic) and physiological profiling presented in this study (top) as well as a table (bottom) highlighting mortality rates at various parts in the parabiosis protocol (numbers represent individual mice, not pairs). Samples were taken immediately after the parabiosis period (month 3), or after a 2-month detachment period (month 5). Physiological profiling was performed before the parabiosis experiment (month 0), and then every month from month 4 to month 10. Data in the table (bottom) is separated by experimental purposes and ages of the pairs. Mortality rate across experiments for each group: Young ISO: 11% mortality during PB, 0% during detach; Old ISO: 20% mortality during PB, 13% during detach. Old HET: 19% mortality during PB, 7% during detach. b, Changes in food consumption resulting from parabiosis. Lines depict mean changes with 95% confidence intervals. Individual observations are shown as points. Young isochronic (Young ISO) mice are shown in blue, old heterochronic (Old HET) mice are shown in purple, and old isochronic (Old ISO) mice are shown in red. Dashed line depicts time of detachment. c, Changes in vertical activity resulting from parabiosis. Lines depict mean changes with 95% confidence intervals. Individual observations are shown as points. Legend is the same as (b). d, Schematic of the experimental workflow and associated findings.

Source data

Extended Data Fig. 2 Blood mixture analyses after 3 months of parabiosis.

a, Schematic of experiments to analyze the blood chimerism after attachment and detachment. Green represents GFP (+) mice, white represents wild-type mice. b, Gating scheme applied to all downstream blood analysis between GFP and wild-type mice. Cells were ultimately gated on GFP (+) (x-axis) and CD45-APC (+) (y-axis). c, Percentage of blood crossover during attachment. d, Percentage of blood (left) and bone marrow (BM, right) crossover after 2-month detachment, calculated as the % GFP (+) and (-) cells, n = 3 biological replicates group. Bars represent mean +/− SEM of cell proportions labelled GFP+ and GFP-(WT).

Source data

Extended Data Fig. 3 Relationship of DNAm age across tissues and platforms, delta age analysis, and effect of surgery.

a, Scatterplots highlighting the association of liver and blood DNAm predictions based on RRBS sequencing across 6 different group: young isochronic (Young ISO), young isochronic detached (Young ISO Det.), old heterochronic (Old HET), old isochronic (Old ISO), old heterochronic detached (Old HET Det.), and old isochronic detached (Old ISO Det.). Each point depicts the mean DNAm prediction for a particular tissue for that group. The clock used for predictions is shown in the top part of each panel. The Pearson correlation (r) is shown in each panel, along with its associated two-tailed P value. Linear regression lines (dark grey) with 95% confidence intervals (light grey) are shown. b, Scatterplots highlighting the association of liver DNAm predictions based on RRBS sequencing and methylation array profiling across 6 different groups (same legend as a). The clock used for RRBS predictions is shown in the top part of each panel. The mouse liver development clock was used for array predictions given that it showed the greatest changes in epigenetic age comparing long-term old heterochronic and isochronic mice. c, Delta age (epigenetic age minus chronological age) of liver samples from old heterochronic mice, old isochronic controls and untreated controls based on universal relative age mammalian, universal log-linear transformed age mammalian, liver, and development liver clocks. Dashed line denotes a delta age of 0 (epigenetic age = chronological age). Points above this line depict age acceleration (epigenetic age > chronological age), while points below this line depict age deceleration (epigenetic age < chronological age). n = 5 per group. d, Epigenetic age of liver samples from 8-month old non-surgical control mice (Young NSC), isochronic detached (Young ISO) and heterochronic detached mice (Young HET), based on the universal relative age mammalian, universal log-linear transformed age mammalian, liver, and liver development clocks. n = 5 per group for parabiosed mice and 6 per group for controls. Box plots represent 25–75 percentile and 1.5x IQR. Two-tailed Welch’s t-tests were used for statistical analyses.

Source data

Extended Data Fig. 4 Mean methylation in array and RRBS methylation profiles.

a, Mean liver methylation assayed by the Mammalian Methylation Array (HorvathMammalMethylChip40), both based on all sites in the array (left) and only sites in CpG islands (right). n = 5 samples per group. b, Mean global methylation (top), promoter methylation (middle), and gene body methylation (bottom) of liver (left) and blood (right) RRBS samples. Mean methylation was assayed across 1,014,243 CpGs (top), 11,842 promoters (middle), and 13,811 gene bodies (bottom). n = 4 samples per group. Box plots represent 25–75 percentile and 1.5x IQR. Two-tailed Welch’s t-test were used for statistical analysis. Schematic tissues in each panel indicate the source of the sample.

Source data

Extended Data Fig. 5 Physiological tests of a mock parabiosis procedure to assess the interaction of parabiosis and exercise.

a, Schematic of the experiment to test blood sharing using glucose levels. Glucose is injected into one mouse and subsequently assessed in both mice in mock parabiosis pairs. b, Blood glucose level changes in pairs starting with a young (left) and old (right) glucose bolus to determine blood sharing; n = 5 biological replicates/group. No statistical test was performed. c, Moving distance in cm (left), and velocity (right) in the old isochronic, young isochronic and heterochronic mock parabiosis pairs. n = 4 biological replicates/ group. Bars represents mean +/− SEM. Pdistance(Young ISO—Old ISO) = 0.034 calculated by two-tailed, Student’s t-test. d, Movement tracking in the old isochronic, young isochronic and heterochronic mock parabiosis pairs.

Source data

Extended Data Fig. 6 Tissue specificity of RRBS epigenomic profiles, promoter methylation changes, and proteomic dynamics in HPB.

a-c, Principal component analysis (PCA) of CpG methylation across 1,014,243 CpG sites (a), 11,842 promoters (b), and 13,811 gene bodies (c) in n = 36 liver samples and n = 32 blood samples, with 6 different groups in both tissues: young isochronic (Young ISO), young isochronic detached (Young ISO Det.), old heterochronic (Old HET), old isochronic (Old ISO), old heterochronic detached (Old HET Det.), and old isochronic detached (Old ISO Det.). In all the PCA plots, tissue of origin is the largest source of variation (44–91%). d, Promoter methylation of Cdc20, Sox30, Mpped1, and Ubl5 across liver RRBS samples. Genes were identified after passing a significance threshold (p < 0.05) when comparing young and old mice, as well as old heterochronic and isochronic mice in both attached and detached groups. n = 6 biological replicates/group. P value’s calculated by two-tailed Welch’s t-tests. Box plots represent Mean, 25–75 quartiles and 1.5x IQR. e, Western blot validation (left) and relative protein expression quantification (right) of Sirt3, Gstt2 and C1qb in mice subjected to parabiosis.

Source data

Extended Data Fig. 7 Interactions between different omics modalities.

a, Correlation of changes across experimental groups and readout types. For each comparison in parentheses (that is Young ISO vs. Old ISO), a z-score was computed for either promoter methylation or gene expression. The Pearson correlation of these z-scores was then determined for genes passing a differential expression/methylation threshold of p < 0.05 for the first (left) group listed in each comparison. b, Heat maps highlighting the concordance of readouts and experimental groups. For each heat map, the z-score of genes that are significantly (p < 0.05) differentially expressed or methylated is shown. Genes are ordered by the z-score in the top group. Color bar on the right denotes z-score. The directional concordance, a measure of how directionally aligned the changes between the two readouts/groups are, is shown at the top of each heat map. 50% concordance represents random changes across groups Two-tailed Benchamini-Hochberg corrected FDR was calculated to compare groups.

Source data

Extended Data Fig. 8 Transcriptomic and epigenetic changes resulting from HPB.

a, Boxplots of RLD-transformed, log-normalized counts of Tert, Dnmt3b, Ly6e, Lmna and Pld1 across 6 groups (from left to right: old short-term isochronic (n = 5), old short-term heterochronic (n = 5), old long-term isochronic (n = 3), old long-term heterochronic (n = 3), old long-term isochronic detached (n = 3), and old long-term heterochronic detached (n = 3). b, Mean promoter methylation of the genes mentioned in (a) across liver RRBS samples across 6 groups (from left to right: young long-term isochronic (n = 6), old long-term isochronic (n = 5), old long-term heterochronic (n = 5), young long-term isochronic detached (n = 6), old long-term isochronic detached (n = 7), and old long-term heterochronic detached (n = 6). Color of the box around each gene signifies directionality (green: gene expression increases in Old HET mice compared to ISO, orange: gene expression decreases in Old HET mice compared to ISO). Despite evident changes in expression patterns for these genes, no significant changes in promoter methylation are observed. N represents biological replicates for each group. P values determined by two-tailed Welch’s t-test. Box plots represent median, 25–75 percentile and 1.5x IQR.

Source data

Extended Data Fig. 9 SASP enrichment and differential expression across long-term and short-term HPB.

a-c, Running enrichment score for the senescence-associated secretory phenotype (SASP) gene set, comparing in (a) old isochronic and old heterochronic mice from the long-term HPB, in (b) old detached isochronic and old detached heterochronic mice from long-term HPB, and in (c) old detached isochronic and old detached heterochronic mice from short-term HPB. The P values for the gene set enrichment, along with the adjusted P value are shown in each panel. Positions of individual genes in the gene set are shown as black bars near the bottom of each panel. Long-term attached HPB shows the greatest negative enrichment for heterochronic samples, followed by detached long-term HPB and attached short-term HPB, respectively. d, Boxplots of RLD-transformed, log-normalized count of five SASP genes across 6 groups (from left to right: old short-term isochronic (n = 5), old short-term heterochronic (n = 5), old long-term isochronic (n = 3), old long-term heterochronic (n = 3), old long-term isochronic detached (n = 3), and old long-term heterochronic detached (n = 3). The log2 fold-change (log2FC) and associated P value are shown as calculated by two-tailed Welch’s test. Box plots represent median, 25–75 percentile and 1.5x IQR.

Source data

Extended Data Fig. 10 STRING network representation of significantly down- and upregulated genes.

a, STRING network representation of significantly downregulated genes in livers of old heterochronic mice (n = 421 genes) compared to isochronic mice immediately after detachment or after a 2-month detachment period (protein-protein interaction q value < 1e-16). Genes were filtered based on directionality and absolute value of the log2FC. b, STRING network representation of significantly upregulated genes in livers of old heterochronic mice (n = 337 genes) compared to isochronic mice immediately after detachment or after a 2-month detachment period (protein-protein interaction q value < 1e-16). Genes were filtered based on directionality and absolute value of the log2FC.

Source data

Supplementary information

Source data

Source Data Fig. 1

Statistical source data.

Source Data Fig. 2

Statistical source data.

Source Data Fig. 3

Statistical source data.

Source Data Fig. 4

Statistical source data.

Source Data Fig. 5

Statistical source data.

Source Data Fig. 6

Statistical source data.

Source Data Fig. 7

Statistical source data.

Source Data Extended Data Fig. 1

Statistical source data.

Source Data Extended Data Fig. 2

Statistical source data.

Source Data Extended Data Fig. 3

Statistical source data.

Source Data Extended Data Fig. 4

Statistical source data.

Source Data Extended Data Fig. 5

Statistical source data.

Source Data Extended Data Fig. 6

Statistical source data.

Source Data Extended Data Fig. 7

Statistical source data.

Source Data Extended Data Fig. 8

Statistical source data.

Source Data Extended Data Fig. 9

Statistical source data.

Source Data Extended Data Fig. 10

Statistical source data.

Source Data Immunoblot Images Extended Data Fig. 6

Unmodified blot images.

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.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Zhang, B., Lee, D.E., Trapp, A. et al. Multi-omic rejuvenation and lifespan extension on exposure to youthful circulation. Nat Aging 3, 948–964 (2023). https://doi.org/10.1038/s43587-023-00451-9

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s43587-023-00451-9

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