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:

A conserved complex lipid signature marks human muscle aging and responds to short-term exercise

Abstract

Studies in preclinical models suggest that complex lipids, such as phospholipids, play a role in the regulation of longevity. However, identification of universally conserved complex lipid changes that occur during aging, and how these respond to interventions, is lacking. Here, to comprehensively map how complex lipids change during aging, we profiled ten tissues in young versus aged mice using a lipidomics platform. Strikingly, from >1,200 unique lipids, we found a tissue-wide accumulation of bis(monoacylglycero)phosphate (BMP) during mouse aging. To investigate translational value, we assessed muscle tissue of young and older people, and found a similar marked BMP accumulation in the human aging lipidome. Furthermore, we found that a healthy-aging intervention consisting of moderate-to-vigorous exercise was able to lower BMP levels in postmenopausal female research participants. Our work implicates complex lipid biology as central to aging, identifying a conserved aging lipid signature of BMP accumulation that is modifiable upon a short-term healthy-aging intervention.

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: Lipid classes measured in mouse tissues.
Fig. 2: BMP accumulation is a cross-tissue lipidomic signature of aging.
Fig. 3: BMP accumulation across tissues in aged mice.
Fig. 4: Age-related changes in lipid biosynthesis across tissues for lipid classes.
Fig. 5: Lysosomal and mitochondrial enzyme activity levels with age and BMP increases with age in an independent mouse cohort.
Fig. 6: BMP accumulation in female and male participants.
Fig. 7: Reduction of BMPs following exercise intervention in humans.

Similar content being viewed by others

Data availability

Lipidomics data are available as Supplementary Information accompanying this article as both summary statistics and processed abundance values (see Supplementary Tables 18). Specifically, the mouse ten-tissue aging lipidome is available in Supplementary Table 1, the female muscle aging lipidome is available in Supplementary Table 4, the man muscle aging lipidome is available in Supplementary Table 5, and the exercise intervention muscle lipidome is available in Supplementary Table 6. Physiological data from the human cohorts have been reported in our previous studies as part of a different analysis29,30. All other data supporting the findings of this study are available either in additional Supplementary tables or from the corresponding authors upon reasonable request.

Code availability

Code supporting the findings of this study is available from the corresponding authors upon reasonable request.

References

  1. Libby, P. et al. Atherosclerosis. Nat. Rev. Dis. Prim. 5, 1–18 (2019).

    Google Scholar 

  2. Knottnerus, S. J. G. et al. Disorders of mitochondrial long-chain fatty acid oxidation and the carnitine shuttle. Rev. Endocr. Metab. Disord. 19, 93–106 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Gao, A. W. et al. A sensitive mass spectrometry platform identifies metabolic changes of life history traits in C. elegans. Sci. Rep. 7, 2408 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  4. Houtkooper, R. H. et al. The metabolic footprint of aging in mice. Sci. Rep. 1, 134 (2011).

    Article  PubMed  PubMed Central  Google Scholar 

  5. Fahy, E., Cotter, D., Sud, M. & Subramaniam, S. Lipid classification, structures and tools. Biochim. Biophys. Acta 1811, 637–647 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Vaz, F. M., Pras-Raves, M., Bootsma, A. H. & van Kampen, A. H. C. Principles and practice of lipidomics. J. Inherit. Metab. Dis. 38, 41–52 (2015).

    Article  CAS  PubMed  Google Scholar 

  7. Wortmann, S. B. et al. Mutations in the phospholipid remodeling gene SERAC1 impair mitochondrial function and intracellular cholesterol trafficking and cause dystonia and deafness. Nat. Genet. 44, 797–802 (2012).

    Article  CAS  PubMed  Google Scholar 

  8. Vreken, P. et al. Defective remodeling of cardiolipin and phosphatidylglycerol in Barth syndrome. Biochem. Biophys. Res. Commun. 279, 378–382 (2000).

    Article  CAS  PubMed  Google Scholar 

  9. Vaz, F. M. et al. Mutations in PCYT2 disrupt etherlipid biosynthesis and cause a complex hereditary spastic paraplegia. Brain 142, 3382–3397 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  10. Ferdinandusse, S. et al. An autosomal dominant neurological disorder caused by de novo variants in FAR1 resulting in uncontrolled synthesis of ether lipids. Genet. Med. 23, 740–750 (2021).

    Article  CAS  PubMed  Google Scholar 

  11. Sustarsic, E. G. et al. Cardiolipin synthesis in brown and beige fat mitochondria is essential for systemic energy homeostasis. Cell Metab. 28, 159–174 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Lynes, M. D. et al. Cold-activated lipid dynamics in adipose tissue highlights a role for cardiolipin in thermogenic metabolism. Cell Rep. 24, 781–790 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Jha, P. et al. Systems analyses reveal physiological roles and genetic regulators of liver lipid species. Cell Syst. 6, 722–733 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Ferrara, P. J. et al. Lysophospholipid acylation modulates plasma membrane lipid organization and insulin sensitivity in skeletal muscle. J. Clin. Invest. 3, e135963 (2021).

  15. Held, N. M. et al. Skeletal muscle in healthy humans exhibits a day-night rhythm in lipid metabolism. Mol. Metab. 37, 100989 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. 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 

  17. Johnson, A. A. & Stolzing, A. The role of lipid metabolism in aging, lifespan regulation, and age-related disease. Aging Cell 18, e13048 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Wang, M. C., O’Rourke, E. J. & Ruvkun, G. Fat metabolism links germline stem cells and longevity in C. elegans. Science 322, 957–960 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Seah, N. E. et al. Autophagy-mediated longevity is modulated by lipoprotein biogenesis. Autophagy 12, 261–272 (2016).

    Article  CAS  PubMed  Google Scholar 

  20. Bozek, K. et al. Lipidome determinants of maximal lifespan in mammals. Sci. Rep. 7, 5 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  21. Akgoc, Z. et al. Bis(monoacylglycero)phosphate: a secondary storage lipid in the gangliosidoses. J. Lipid Res. 56, 1006–1013 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Thornburg, T., Miller, C., Thuren, T., King, L. & Waite, M. Glycerol reorientation during the conversion of phosphatidylglycerol to bis(monoacylglycerol)phosphate in macrophage-like RAW 264.7 cells. J. Biol. Chem. 266, 6834–6840 (1991).

  23. Brotherus, J., Renkonen, O., Herrmann, J. & Fischer, W. Novel stereoconfiguration in lyso-bis-phosphatidic acid of cultured BHK-cells. Chem. Phys. Lipids 13, 178–182 (1974).

    Article  CAS  PubMed  Google Scholar 

  24. Showalter, M. R. et al. The emerging and diverse roles of bis(monoacylglycero) phosphate lipids in cellular physiology and disease. Int. J. Mol. Sci. 21, 8067 (2020).

  25. Medoh, U. N. et al. The Batten disease gene product CLN5 is the lysosomal bis(monoacylglycero)phosphate synthase. Science 381, 1182–1189 (2023).

    Article  CAS  PubMed  Google Scholar 

  26. Herzog, K. et al. Lipidomic analysis of fibroblasts from Zellweger spectrum disorder patients identifies disease-specific phospholipid ratios. J. Lipid Res. 57, 1447–1454 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Chen, J. et al. Lysosomal phospholipase A2 contributes to the biosynthesis of the atypical late endosome lipid bis(monoacylglycero)phosphate. Commun. Biol. 6, 2–10 (2023).

    Article  PubMed  PubMed Central  Google Scholar 

  28. Held, N. M. et al. Aging selectively dampens oscillation of lipid abundance in white and brown adipose tissue. Sci. Rep. 11, 5932 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Grevendonk, L. et al. Impact of aging and exercise on skeletal muscle mitochondrial capacity, energy metabolism, and physical function. Nat. Commun. 12, 4773 (2021).

  30. Remie, C. M. E. et al. Sitting less elicits metabolic responses similar to exercise and enhances insulin sensitivity in postmenopausal women. Diabetologia 64, 2817–2828 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Grabner, G. F. et al. Metabolic regulation of the lysosomal cofactor bis(monoacylglycero)phosphate in mice. J. Lipid Res. 61, 995–1003 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Rampanelli, E. et al. Metabolic injury-induced NLRP3 inflammasome activation dampens phospholipid degradation. Sci. Rep. 7, 2861 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  33. Luquain-Costaz, C. et al. Bis(monoacylglycero)phosphate accumulation in macrophages induces intracellular cholesterol redistribution, attenuates liver-X receptor/ATP-binding cassette transporter A1/ATP-binding cassette transporter G1 pathway, and impairs cholesterol efflux. Arterioscler. Thromb. Vasc. Biol. 33, 1803–1811 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Hullin-Matsuda, F., Luquain-Costaz, C., Bouvier, J. & Delton-Vandenbroucke, I. Bis(monoacylglycero)phosphate, a peculiar phospholipid to control the fate of cholesterol: implications in pathology. Prostaglandins Leukot. Essent. Fatty Acids 81, 313–324 (2009).

  35. Meikle, P. J. et al. Effect of lysosomal storage on bis(monoacylglycero)phosphate. Biochem. J. 411, 71–78 (2008).

    Article  CAS  PubMed  Google Scholar 

  36. Schuurman, A. R. et al. The platelet lipidome is altered in patients with COVID-19 and correlates with platelet reactivity. Thromb. Haemost. 122, 1683–1692 (2022).

    Article  PubMed  PubMed Central  Google Scholar 

  37. Falabella, M., Vernon, H. J., Hanna, M. G., Claypool, S. M. & Pitceathly, R. D. S. Cardiolipin, mitochondria, and neurological disease. Trends Endocrinol. Metab. 32, 224–237 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Liu, D., Aziz, N. A., Pehlivan, G. & Breteler, M. M. B. Lipidomic correlates of epigenetic aging across the adult lifespan: a population-based study. GeroScience https://doi.org/10.1007/s11357-022-00714-0 (2023).

  39. Cedillo, L. et al. Ether lipid biosynthesis promotes lifespan extension and enables diverse pro-longevity paradigms in Caenorhabditis elegans. eLife 12, e82210 (2023).

  40. Baars, A. et al. Sex differences in lipid metabolism are affected by presence of the gut microbiota. Sci. Rep. 8, 13426 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  41. González-Granillo, M. et al. Sex-specific lipid molecular signatures in obesity-associated metabolic dysfunctions revealed by lipidomic characterization in ob/ob mouse. Biol. Sex Differ. 10, 11 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  42. Holcomb, L. E., Rowe, P., O’Neill, C. C., DeWitt, E. A. & Kolwicz, S. C. Sex differences in endurance exercise capacity and skeletal muscle lipid metabolism in mice. Physiol. Rep. 10, (2022).

  43. Mouchiroud, L. et al. The NAD+/sirtuin pathway modulates longevity through activation of mitochondrial UPR and FOXO signaling. Cell 154, 430–441 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  44. Smith, P. K. et al. Measurement of protein using bicinchoninic acid. Anal. Biochem. 150, 76–85 (1985).

    Article  CAS  PubMed  Google Scholar 

  45. Chambers, M. C. et al. A cross-platform toolkit for mass spectrometry and proteomics. Nat. Biotechnol. 30, 918–920 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. R Core Team. A language and environment for statistical computing. R Foundation for Statistical Computing https://www.r-project.org/ (2019).

  47. Smith, C. A., Want, E. J., O’Maille, G., Abagyan, R. & Siuzdak, G. XCMS: processing mass spectrometry data for metabolite profiling using nonlinear peak alignment, matching, and identification. Anal. Chem. 78, 779–787 (2006).

  48. Ruiz, M. et al. Lipidomics unveils lipid dyshomeostasis and low circulating plasmalogens as biomarkers in a monogenic mitochondrial disorder. JCI Insight 4, e123231 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  49. Huffnagel, I. C. et al. Disease progression in women with X-linked adrenoleukodystrophy is slow. Orphanet J. Rare Dis. https://doi.org/10.1186/s13023-019-1008-6 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  50. Bergström, J. Muscle electrolytes in man determined by neutron activation analysis on needle biopsy specimens. Scand. J. Clin. Lab. Invest. 14, 7–110 (1962).

    Google Scholar 

  51. Molenaars, M. et al. Metabolomics and lipidomics in C. elegans using a single sample preparation. Dis. Model. Mech. https://doi.org/10.1242/dmm.047746 (2021).

    Article  PubMed  PubMed Central  Google Scholar 

  52. Gentleman, R. C. et al. Bioconductor: open software development for computational biology and bioinformatics. Genome Biol. 5, R80 (2004).

    Article  PubMed  PubMed Central  Google Scholar 

  53. Rohart, F., Gautier, B., Singh, A. & Lê Cao, K. A. mixOmics: an R package for ‘omics feature selection and multiple data integration. PLoS Comput. Biol. 13, e1005752 (2017).

  54. Wickham, H. Ggplot2. Wiley Interdiscip. Rev. Comput. Stat. 3, 180–185 (2011).

    Article  Google Scholar 

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

    Article  PubMed  PubMed Central  Google Scholar 

  56. Law, C. W., Chen, Y., Shi, W. & Smyth, G. K. voom: precision weights unlock linear model analysis tools for RNA-seq read counts. Genome Biol. 15, R29 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

Work in the R.H.H. group is financially supported by an ERC starting grant (no. 638290), by a VIDI grant from ZonMw (no. 91715305) and by the Velux Stiftung (no. 1063). G.E.J. is supported by a VENI grant from ZonMw, and AGEM Talent and Development grants. The project was supported by a Longevity Impetus Grant from Norn Group (to G.E.J. and R.H.H.). Human interventions were further financed by the TIFN research program Mitochondrial Health (ALWTF.2015.5) and the Netherlands Organization for Scientific Research (NWO). The funders had no role in data collection, analysis or decision to publish or in preparation of the paper.

Author information

Authors and Affiliations

Authors

Contributions

G.E.J. and R.H.H. conceived and designed lipidomics experiments and analyses; R.H.H. designed and performed mouse experiments; L.G., J.H., C.M.E.R. and P.S. designed and performed human experiments; G.E.J., K.H., M.A.T.V., B.V.S., H.L.E., E.J.M.W., P.D.M., H.R.W., M.L.P.-R., M.v.W., A.H.C.v.K., F.M.V., A.J., A.T., L.M.B., S.F. and M.M. performed lipidomics sample preparation, data generation, bioinformatics or consultation; S.W.D. and S.v.d.R. performed enzymatic assays and statistics; G.E.J. designed and performed analyses and integration of all datasets; G.E.J. and R.H.H. interpreted the data and wrote the paper with contributions from all authors.

Corresponding authors

Correspondence to Georges E. Janssens, Frédéric M. Vaz or Riekelt H. Houtkooper.

Ethics declarations

Competing interests

The authors declare no competing interests.

Peer review

Peer review information

Nature Aging thanks Luigi Ferrucci, Jørn Wulff Helge and the other, anonymous, reviewer(s) 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 Characteristics of cohorts used in this study.

Cohorts are shown for both the Aging study (pertaining to Fig. 6) and Sitless study (pertaining to Fig. 7). * Values are averages (mean) ± standard deviation (SD). Sex data are presented as the number of participants. Source data can be found in the original study cohort descriptions, see Grevendonk et al.29 and Remie et al.30.

Extended Data Fig. 2 Comparison of lipid changes in muscle biopsies of aging men and women.

a, Principal component analysis (PCA) of muscle lipidomes of either men (top panel) or women (bottom panel) from this study. b, correlation between the fold change of lipids induced by aging in either women (y-axis) or men (x-axis) shows significant correlation of age-related lipid changes (r = 0.53, p = 1.66e-111, Pearson’s correlation). c, Volcano plot highlighting significantly altered BMP species in men reveals accumulation of many BMP lipid species. The horizontal line indicates significance (p < 0.05) where significance was determined using an empirical Bayes moderated t-test (two sided). d, Fisher′s exact test (one sided) showing enrichment of individual BMPs in the significantly accumulated fraction of lipids (p = 1e-4), pertaining to panel c. e, comparison of lipid changes between men and women where changes are evaluated as p values in –log10 scale, multiplied by the sign of the fold change (such that depleted lipids are having a negative value and accumulating lipids a positive value). BMPs significantly accumulated in both men and women are highlighted in red. Significance was determined using a Student’s t test (two sided). f, Top Venn diagram: overlap of significantly accumulated lipids in men and women, used for bottom Venn diagram, left circle. Bottom Venn diagram: overlap of lipids accumulated in both men and women and BMPs, used for Fisher’s exact test (one sided). Reveals enrichment of BMPs in the fraction of lipids accumulating in common between men and women (p = 2e-05). g-h, Assessment of all lipid classes for enrichment of lipid species in the depleted fraction of changes (Fisher′s exact test, one sided) in g, women and h, men. Corresponds to main Fig. 3d,e. For source data file see: Supplementary Data Table 4 (women) and 5 (men).

Extended Data Fig. 3 Human intervention lipidome with and without randomization scheme in statistical analysis.

Assessment of the human intervention lipidome at the lipid class level, both without (x axis) and with (y axis) taking into account the order in which the participants underwent a regimen. Scatterplots compare the fold changes (log2 scale) of a, exercise vs sit, b, sitless vs sit, and c, exercise vs sitless derived from both statistical comparisons (where significance was determined using an empirical Bayes moderated t-test, two sided). High Pearson’s correlations observed between both statistical approaches (>0.9) and significance values (p < e-10) indicates that the order in which a participant underwent an intervention did not greatly impact the lipidomics analyses. For Source Data file see Supplementary Data Table 7.

Supplementary information

Supplementary Information

Lipidomicstandard.org reporting summaries for (1) aging mouse, (2) aging human (man and woman) and (3) exercise intervention lipidomics data.

Reporting Summary

Supplementary Table 1

Mouse aging lipidomics data (for Fig. 1 and Extended Data Fig. 1).

Supplementary Table 2

Mouse pathway lipidomics data (for Fig. 32).

Supplementary Table 3

Mouse enzyme assay data (for Extended Data Fig. 3).

Supplementary Table 4

Female aging lipidomics data (for Fig. 3 and Extended Data Fig. 5).

Supplementary Table 5

Male aging lipidomics data (also for Fig. 3 and Extended Data Fig. 5)

Supplementary Table 6

Exercise intervention lipidomics data (for Fig. 4).

Supplementary Table 7

Exercise intervention washout comparison (for Extended Data Fig. 6).

Supplementary Table 8

Pooled control in lipidomics with coefficient of variation measures per lipid species (for Methods section).

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

Janssens, G.E., Molenaars, M., Herzog, K. et al. A conserved complex lipid signature marks human muscle aging and responds to short-term exercise. Nat Aging (2024). https://doi.org/10.1038/s43587-024-00595-2

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1038/s43587-024-00595-2

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