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:

Regulation of aged skeletal muscle regeneration by circulating extracellular vesicles

Abstract

Heterochronic blood exchange (HBE) has demonstrated that circulating factors restore youthful features to aged tissues. However, the systemic mediators of those rejuvenating effects remain poorly defined. We show here that the beneficial effect of young blood on aged muscle regeneration was diminished when serum was depleted of extracellular vesicles (EVs). Whereas EVs from young animals rejuvenate aged cell bioenergetics and skeletal muscle regeneration, aging shifts EV subpopulation heterogeneity and compromises downstream benefits on recipient cells. Machine learning classifiers revealed that aging shifts the nucleic acid, but not protein, fingerprint of circulating EVs. Alterations in subpopulation heterogeneity were accompanied by declines in transcript levels of the prolongevity protein α-Klotho (Klotho), and injection of EVs improved muscle regeneration in a Klotho mRNA-dependent manner. These studies demonstrate that EVs play a key role in the rejuvenating effects of HBE and that Klotho transcripts within EVs phenocopy the effects of young serum on aged skeletal muscle.

This is a preview of subscription content, access via your institution

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

Prices may be subject to local taxes which are calculated during checkout

Fig. 1: The beneficial effect of young serum on aged muscle progenitors is dependent on circulating EVs.
Fig. 2: The beneficial effect of young serum on aged muscle regeneration and mitochondrial function is dependent, at least in part, on circulating EVs.
Fig. 3: Aging shifts EV subpopulation heterogeneity and disrupts the biochemical fingerprint of EVs.
Fig. 4: Transcriptomic alterations in skeletal muscle with young serum treatment are dominated by EVs.
Fig. 5: Klotho transcripts in EVs decline over time and are preferentially contained within EVs with high expression of the CD81 surface marker.
Fig. 6: EV age impacts skeletal muscle regeneration and function.
Fig. 7: Klotho mRNA within EVs contributes to functional skeletal muscle regeneration.

Similar content being viewed by others

Data availability

The raw data that support the experimental findings are included as Supplementary Information. The image files used for computational analysis are available at https://github.com/ankitbhatia/bioimage_aging. RNA-sequencing data have been deposited to the NCBI Gene Expression Omnibus database with the accession number GSE176478.

Code availability

The code used to perform machine learning-based analyses of EVs is available at https://github.com/ankitbhatia/bioimage_aging. For PC analyses, the OriginLab plugin ‘Principal Component Analysis for Spectroscopy’ was used. The code for RNA-sequencing analysis is available at https://github.com/sruthi-hub/Aging_EV/tree/main.

References

  1. 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 

  2. 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 

  3. Brack, A. S. et al. Increased Wnt signaling during aging alters muscle stem cell fate and increases fibrosis. Science 317, 807–810 (2007).

    Article  CAS  PubMed  Google Scholar 

  4. Sousa-Victor, P. et al. MANF regulates metabolic and immune homeostasis in ageing and protects against liver damage. Nat. Metab. 1, 276–290 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Li, L. et al. Impairment of chondrocyte proliferation after exposure of young murine cartilage to an aged systemic environment in a heterochronic parabiosis model. Swiss Med. Wkly. 148, w14607 (2018).

    PubMed  Google Scholar 

  6. Gontier, G. et al. Tet2 rescues age-related regenerative decline and enhances cognitive function in the adult mouse brain. Cell Rep. 22, 1974–1981 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Pathan, M. et al. Vesiclepedia 2019: a compendium of RNA, proteins, lipids and metabolites in extracellular vesicles. Nucleic Acids Res. 47, D516–D519 (2019).

    Article  CAS  PubMed  Google Scholar 

  8. Shah, R., Patel, T. & Freedman, J. E. Circulating extracellular vesicles in human disease. N. Engl. J. Med. 379, 2180–2181 (2018).

    Article  PubMed  Google Scholar 

  9. Chen, W. W. et al. BEAMing and DropletDigital PCR analysis of mutant IDH1 mRNA in glioma patient serum and cerebrospinal fluid extracellular vesicles. Mol. Ther. Nucleic Acids 2, e109 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  10. Yang, J., Wei, F., Schafer, C. & Wong, D. T. Detection of tumor cell-specific mRNA and protein in exosome-like microvesicles from blood and saliva. PLoS ONE 9, e110641 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  11. Salih, M., Zietse, R. & Hoorn, E. J. Urinary extracellular vesicles and the kidney: biomarkers and beyond. Am. J. Physiol. Renal Physiol. 306, F1251–F1259 (2014).

    Article  CAS  PubMed  Google Scholar 

  12. Monguio-Tortajada, M. et al. Extracellular-vesicle isolation from different biological fluids by size-exclusion chromatography. Curr. Protoc. Stem Cell Biol. 49, e82 (2019).

    PubMed  Google Scholar 

  13. Arraud, N. et al. Extracellular vesicles from blood plasma: determination of their morphology, size, phenotype and concentration. J. Thromb. Haemost. 12, 614–627 (2014).

    Article  CAS  PubMed  Google Scholar 

  14. Revenfeld, A. L. et al. Diagnostic and prognostic potential of extracellular vesicles in peripheral blood. Clin. Ther. 36, 830–846 (2014).

    Article  PubMed  Google Scholar 

  15. Valadi, H. et al. Exosome-mediated transfer of mRNAs and microRNAs is a novel mechanism of genetic exchange between cells. Nat. Cell Biol. 9, 654–659 (2007).

    Article  CAS  PubMed  Google Scholar 

  16. Robbins, P. D. Extracellular vesicles and aging. Stem Cell Investig. 4, 98 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  17. Alibhai, F. J. et al. Cellular senescence contributes to age-dependent changes in circulating extracellular vesicle cargo and function. Aging Cell 19, e13103 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Yoshida, M. et al. Extracellular vesicle-contained eNAMPT delays aging and extends lifespan in mice. Cell Metab. 30, 329–342 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Picca, A. et al. Mitochondrial dysfunction and aging: insights from the analysis of extracellular vesicles. Int. J. Mol. Sci. 20, 805 (2019).

    Article  CAS  PubMed Central  Google Scholar 

  20. Dubal, D. B. et al. Life extension factor Klotho enhances cognition. Cell Rep. 7, 1065–1076 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Sahu, A. et al. Age-related declines in alpha-Klotho drive progenitor cell mitochondrial dysfunction and impaired muscle regeneration. Nat. Commun. 9, 4859 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Ahrens, H. E., Huettemeister, J., Schmidt, M., Kaether, C. & von Maltzahn, J. Klotho expression is a prerequisite for proper muscle stem cell function and regeneration of skeletal muscle. Skelet Muscle 8, 20 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  23. Tapscott, S. J. The circuitry of a master switch: Myod and the regulation of skeletal muscle gene transcription. Development 132, 2685–2695 (2005).

    Article  CAS  PubMed  Google Scholar 

  24. Zammit, P. S. et al. Pax7 and myogenic progression in skeletal muscle satellite cells. J. Cell Sci. 119, 1824–1832 (2006).

    Article  CAS  PubMed  Google Scholar 

  25. Zhang, H. et al. NAD+ repletion improves mitochondrial and stem cell function and enhances life span in mice. Science 352, 1436–1443 (2016).

    Article  CAS  PubMed  Google Scholar 

  26. Pala, F. et al. Distinct metabolic states govern skeletal muscle stem cell fates during prenatal and postnatal myogenesis. J. Cell Sci. 131, jcs212977 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  27. Paradies, G., Paradies, V., De Benedictis, V., Ruggiero, F. M. & Petrosillo, G. Functional role of cardiolipin in mitochondrial bioenergetics. Biochim. Biophys. Acta 1837, 408–417 (2014).

    Article  CAS  PubMed  Google Scholar 

  28. Cecchini, G. Function and structure of complex II of the respiratory chain. Annu. Rev. Biochem. 72, 77–109 (2003).

    Article  CAS  PubMed  Google Scholar 

  29. Gamez-Valero, A. et al. Size-exclusion chromatography-based isolation minimally alters extracellular vesicles’ characteristics compared to precipitating agents. Sci. Rep. 6, 33641 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Andreu, Z. & Yanez-Mo, M. Tetraspanins in extracellular vesicle formation and function. Front. Immunol. 5, 442 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  31. Moon, S. Enrichment of exosome-like extracellular vesicles from plasma suitable for clinical vesicular miRNA biomarker research. J. Clin. Med. 8, 1995 (2019).

    Article  CAS  PubMed Central  Google Scholar 

  32. 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 

  33. Thery, C. et al. Minimal information for studies of extracellular vesicles 2018 (MISEV2018): a position statement of the International Society for Extracellular Vesicles and update of the MISEV2014 guidelines. J. Extracell. Vesicles 7, 1535750 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  34. Mastoridis, S. et al. Multiparametric analysis of circulating exosomes and other small extracellular vesicles by advanced imaging flow cytometry. Front. Immunol. 9, 1583 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  35. Gualerzi, A. et al. Raman spectroscopy uncovers biochemical tissue-related features of extracellular vesicles from mesenchymal stromal cells. Sci. Rep. 7, 9820 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  36. Movasaghi Z., Rehman S., & Rehman I.U., Raman spectroscopy of biological tissues. Appl. Spectrosc. Rev. 42, 493–541 (2007).

    Article  Google Scholar 

  37. de Cavanagh, E. M. V., Inserra, F., & Ferder, L., Angiotensin II blockade: how its molecular targets may signal to mitochondria and slow aging. Coincidences with calorie restriction and mTOR inhibition. Am. J. Physiol. Heart Circ. Physiol. 309, H15–H44 (2015).

    Article  Google Scholar 

  38. Murphy, E. & Eisner, D. A. Regulation of intracellular and mitochondrial sodium in health and disease. Circ. Res. 104, 292–303 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Brookes, P. S., Yoon, Y., Robotham, J. L., Anders, M. W. & Sheu, S. S. Calcium, ATP, and ROS: a mitochondrial love-hate triangle. Am. J. Physiol. Cell Physiol. 287, C817–C833 (2004).

    Article  CAS  PubMed  Google Scholar 

  40. Garlid, K. D. & Paucek, P. Mitochondrial potassium transport: the K+ cycle. Biochim. Biophys. Acta 1606, 23–41 (2003).

    Article  CAS  PubMed  Google Scholar 

  41. Garth, J. et al. The effects of the anti-aging protein Klotho on mucociliary clearance. Front. Med. 6, 339 (2019).

    Article  Google Scholar 

  42. Tang, G., Shen, Y., Gao, P., Song, S. S. & Si, L. Y. Klotho attenuates isoproterenol-induced hypertrophic response in H9C2 cells by activating Na(+)/K(+)-ATPase and inhibiting the reverse mode of Na+/Ca2+-exchanger. In Vitro Cell Dev. Biol. Anim. 54, 250–256 (2018).

    Article  CAS  PubMed  Google Scholar 

  43. Shumilina, E. et al. Altered regulation of cytosolic Ca2+ concentration in dendritic cells from klotho hypomorphic mice. Am. J. Physiol. Cell Physiol. 305, C70–C77 (2013).

    Article  CAS  PubMed  Google Scholar 

  44. Strutz-Seebohm, N., Wrobel, E., Schulze-Bahr, E. & Seebohm, G. Klotho: a new trafficking modifier of Kv7.1/KCNE1 channels. Channels 8, 285 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  45. Ohnishi, M. & Razzaque, M. S. Dietary and genetic evidence for phosphate toxicity accelerating mammalian aging. FASEB J. 24, 3562–3571 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Wehling-Henricks, M. et al. Klotho gene silencing promotes pathology in the mdx mouse model of Duchenne muscular dystrophy. Hum. Mol. Genet. 25, 2465–2482 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  47. Cheikhi, A. et al. Klotho: an elephant in aging research. J. Gerontol. A Biol. Sci. Med. Sci. 74, 1031–1042 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Gonzales, P. A. et al. Large-scale proteomics and phosphoproteomics of urinary exosomes. J. Am. Soc. Nephrol. 20, 363–379 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Skog, J. et al. Glioblastoma microvesicles transport RNA and proteins that promote tumour growth and provide diagnostic biomarkers. Nat. Cell Biol. 10, 1470–1476 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Zomer, A. et al. In vivo imaging reveals extracellular vesicle-mediated phenocopying of metastatic behavior. Cell 161, 1046–1057 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. de la Cuesta, F. et al. Extracellular vesicle cross-talk between pulmonary artery smooth muscle cells and endothelium during excessive TGF-beta signalling: implications for PAH vascular remodelling. Cell Commun. Signal. 17, 143 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  52. Kuro-o, M. et al. Mutation of the mouse Klotho gene leads to a syndrome resembling ageing. Nature 390, 45–51 (1997).

    Article  CAS  PubMed  Google Scholar 

  53. Li, S. et al. exoRBase: a database of circRNA, lncRNA and mRNA in human blood exosomes. Nucleic Acids Res. 46, D106–D112 (2018).

    Article  CAS  PubMed  Google Scholar 

  54. Egerman, M. A. et al. GDF11 increases with age and inhibits skeletal muscle regeneration. Cell Metab. 22, 164–174 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Choi, J. S. et al. Exosomes from differentiating human skeletal muscle cells trigger myogenesis of stem cells and provide biochemical cues for skeletal muscle regeneration. J. Control. Release 222, 107–115 (2016).

    Article  CAS  PubMed  Google Scholar 

  56. Eldh, M. et al. Exosomes communicate protective messages during oxidative stress: possible role of exosomal shuttle RNA. PLoS ONE 5, e15353 (2010).

    Article  PubMed  PubMed Central  Google Scholar 

  57. Fry, C. S., Kirby, T. J., Kosmac, K., McCarthy, J. J. & Peterson, C. A. Myogenic progenitor cells control extracellular matrix production by fibroblasts during skeletal muscle hypertrophy. Cell Stem Cell 20, 56–69 (2017).

    Article  CAS  PubMed  Google Scholar 

  58. Villarroya-Beltri, C. et al. Sumoylated hnRNPA2B1 controls the sorting of miRNAs into exosomes through binding to specific motifs. Nat. Commun. 4, 2980 (2013).

    Article  PubMed  Google Scholar 

  59. de Moura, M. B. & Van Houten, B. Bioenergetic analysis of intact mammalian cells using the Seahorse XF24 Extracellular Flux analyzer and a luciferase ATP assay. Methods Mol. Biol. 1105, 589–602 (2014).

    Article  PubMed  Google Scholar 

  60. Zhang, C. et al. Arsenic promotes NF-kappaB-mediated fibroblast dysfunction and matrix remodeling to impair muscle stem cell function. Stem Cells 34, 732–742 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

These studies were supported by NIA grants R01AG052978 (F.A.), R01AG061005 (F.A.), R01AG066198-01 (F.A. and R.K.) and R33 ES025606-05 (B.V.H.) and UPMC Enterprises (F.A.). We thank the flow cytometry core at the Department of Immunology, University of Pittsburgh for providing resources and expertise to perform ImageStream analysis (National Institutes of Health grant 1S10OD019942-01), as well as the Center of Biologic Imaging, University of Pittsburgh for providing resources to perform confocal imaging (National Institutes of Health grant 1S10OD019973). The funders had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript.

Author information

Authors and Affiliations

Authors

Contributions

F.A. and A.S. provided the concept, idea and experimental design for the studies. F.A. and A.S. wrote the manuscript. A.S., Z.J.C., S.S., S.N.S. and A.P. collected, analyzed and interpreted data and reviewed the manuscript. A. Bhatia provided computer vision- and machine learning-based analyses. S.P., C.C., and A.G. collected and analyzed data. M.B. interpreted data and reviewed the manuscript. B.V.H. analyzed and interpreted data and reviewed that manuscript. A. Barchowsky provided consultation for data interpretation and review of the manuscript. M.L. provided support for statistical analyses. N.F., I.L., and R.K. provided support for data interpretation. F.A. provided funding for the studies.

Corresponding author

Correspondence to Fabrisia Ambrosio.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Peer review information Nature Aging thanks Dan Lark, Tim Gavin, and the other, 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 Depletion of EVs eliminates the effect of young serum on Pax7 expression of muscle progenitors.

Quantification of Pax7 in aged MPCs treated with aged serum, young serum or EV-depleted aged or young serum. Scale bars, 50 µm (****P < 0.0001, two-tailed Mann–Whitney test comparing depleted young serum and young serum treatments). Data are presented as mean + s.e.m. Data from different cohorts or experimental groups performed on different days are presented within the same graph as black or red circles.

Source data

Extended Data Fig. 2 The ability of EVs to modulate target cell Klotho and MyoD protein levels is dependent on Klotho mRNAs.

a, Imaging and quantification of Klotho protein in aged MPCs following culture in the presence of young or aged EVs for 24 h. Scale bars, 50 µm (n = 6 wells per group performed over two independent experiments, **P < 0.01, two-tailed Welch’s t test). b, Representative violin plot of Klotho protein intensity per EV from young and aged serum, using imaging flow cytometry (n = 11,229–11,685 EVs per group for this experimental run. EVs pooled from four young and four aged serum samples, P > 0.05, two-tailed Mann–Whitney test, experiment repeated in triplicate). Violin plot minima, maxima, median, 25th percentile and 75th percentile are 0, 272915.9, 0, 0 and 26.36 (young serum) and 0, 272241.3, 0, 0, and 23.2 (aged serum). c,d, Quantification of MyoD-positive (%), desmin-positive (%) and ki67-positive (%) aged MPCs receiving young serum EVs treated with scramble or siRNA to Klotho (c) or aged serum EVs or aged serum EVs loaded with synthetic Klotho mRNA (d) (MyoD and desmin (scramble, siRNA) or desmin (aged EVs, synthetic Klotho): **P < 0.01, ***P < 0.001 and ****P < 0.0001, two-tailed t test with Welch’s correction, n = 5–6 wells per group; MyoD (aged EVs and synthetic Klotho): **P < 0.01, two-tailed Mann–Whitney test, n = 5 wells and group; ki67 (scramble, siRNA and aged EVs and synthetic Klotho): P > 0.05 (P = 0.1), two-tailed Mann–Whitney test).

Source data

Supplementary information

Source data

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Sahu, A., Clemens, Z.J., Shinde, S.N. et al. Regulation of aged skeletal muscle regeneration by circulating extracellular vesicles. Nat Aging 1, 1148–1161 (2021). https://doi.org/10.1038/s43587-021-00143-2

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s43587-021-00143-2

Search

Quick links

Nature Briefing: Translational Research

Sign up for the Nature Briefing: Translational Research newsletter — top stories in biotechnology, drug discovery and pharma.

Get what matters in translational research, free to your inbox weekly. Sign up for Nature Briefing: Translational Research