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:

Eosinophils regulate adipose tissue inflammation and sustain physical and immunological fitness in old age

Abstract

Adipose tissue eosinophils (ATEs) are important in the control of obesity-associated inflammation and metabolic disease. However, the way in which ageing impacts the regulatory role of ATEs remains unknown. Here, we show that ATEs undergo major age-related changes in distribution and function associated with impaired adipose tissue homeostasis and systemic low-grade inflammation in both humans and mice. We find that exposure to a young systemic environment partially restores ATE distribution in aged parabionts and reduces adipose tissue inflammation. Approaches to restore ATE distribution using adoptive transfer of eosinophils from young mice into aged recipients proved sufficient to dampen age-related local and systemic low-grade inflammation. Importantly, restoration of a youthful systemic milieu by means of eosinophil transfers resulted in systemic rejuvenation of the aged host, manifesting in improved physical and immune fitness that was partially mediated by eosinophil-derived IL-4. Together, these findings support a critical function of adipose tissue as a source of pro-ageing factors and uncover a new role of eosinophils in promoting healthy ageing by sustaining adipose tissue homeostasis.

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: Age-related changes in the plasma proteome of humans and mice.
Fig. 2: Age-related changes in innate immune cell distribution of WAT in humans and mice.
Fig. 3: Heterochronic parabiosis restores ATE/ATM ratios and limits WAT inflammation.
Fig. 4: Transfer of eosinophils from young donors reverses ageing signatures in WAT and limits systemic inflammation.
Fig. 5: Eosinophils from young donors improve physical fitness in aged hosts.
Fig. 6: Eosinophil homing to WAT lowers age-related adipose tissue inflammation and improves physical fitness in aged recipients.
Fig. 7: Transfer of eosinophils into aged mice transiently alters HSC numbers and age-related myeloid skewing.
Fig. 8: Transfer of eosinophils into aged mice is associated with improved immunological fitness.
Fig. 9: Schematic representation illustrating the rejuvenating potential of young donor eosinophils in the aged host.

Similar content being viewed by others

Data availability

Additional data that support the findings of this study are available from the corresponding authors upon reasonable request. Source data for Extended Data Fig. 1 are available online.

References

  1. Palmer, A. K. & Kirkland, J. L. Aging and adipose tissue: potential interventions for diabetes and regenerative medicine. Exp. Gerontol. 86, 97–105 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Barzilai, N., Banerjee, S., Hawkins, M., Chen, W. & Rossetti, L. Caloric restriction reverses hepatic insulin resistance in aging rats by decreasing visceral fat. J. Clin. Invest. 101, 1353–1361 (1998).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Finkel, T. The metabolic regulation of aging. Nat. Med. 21, 1416–1423 (2015).

    Article  CAS  PubMed  Google Scholar 

  4. López-Otín, C., Galluzzi, L., Freije, J. M. P., Madeo, F. & Kroemer, G. Metabolic control of longevity. Cell 166, 802–821 (2016).

    Article  PubMed  CAS  Google Scholar 

  5. Gabriely, I. et al. Removal of visceral fat prevents insulin resistance and glucose intolerance of aging: an adipokine-mediated process? Diabetes 51, 2951–2958 (2002).

    Article  CAS  PubMed  Google Scholar 

  6. Muzumdar, R. et al. Visceral adipose tissue modulates mammalian longevity. Aging Cell. 7, 438–440 (2008).

    Article  CAS  PubMed  Google Scholar 

  7. Fontana, L., Partridge, L. & Longo, V. D. Extending healthy life span—from yeast to humans. Science 328, 321–326 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Colman, R. J. et al. Caloric restriction delays disease onset and mortality in rhesus monkeys. Science 325, 201–204 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Mattison, J. A. et al. Caloric restriction improves health and survival of rhesus monkeys. Nat. Commun. 8, 14063 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Weindruch, R. The retardation of aging by caloric restriction: studies in rodents and primates. Toxicol. Pathol. 24, 742–745 (1996).

    Article  CAS  PubMed  Google Scholar 

  11. Xu, M. et al. Senolytics improve physical function and increase lifespan in old age. Nat. Med. 55, 1246–1256 (2018).

    Article  CAS  Google Scholar 

  12. Hahn, O. et al. A nutritional memory effect counteracts the benefits of dietary restriction in old mice. Nat. Metab. 1, 1–29 (2019).

    Article  CAS  Google Scholar 

  13. Lumeng, C. N. et al. Aging is associated with an increase in T cells and inflammatory macrophages in visceral adipose tissue. J. Immunol. 187, 6208–6216 (2011).

    Article  CAS  PubMed  Google Scholar 

  14. Rosenberg, H. F., Dyer, K. D. & Foster, P. S. Eosinophils: changing perspectives in health and disease. Nat. Rev. Immunol. 13, 9–22 (2013).

    Article  CAS  PubMed  Google Scholar 

  15. Wu, D. et al. Eosinophils sustain adipose alternatively activated macrophages associated with glucose homeostasis. Science 332, 243–247 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Lackey, D. E. & Olefsky, J. M. Regulation of metabolism by the innate immune system. Nat. Rev. Endocrinol. 12, 15–28 (2016).

    Article  CAS  PubMed  Google Scholar 

  17. Pérez, L. M. et al. ‘Adipaging’: ageing and obesity share biological hallmarks related to a dysfunctional adipose tissue. J. Physiol. 594, 3187–3207 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  18. 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 

  19. Franceschi, C. & Campisi, J. Chronic inflammation (inflammaging) and its potential contribution to age-associated diseases. J. Gerontol. A 69, S4–S9 (2014).

    Article  Google Scholar 

  20. Furman, D. et al. Chronic inflammation in the etiology of disease across the life span. Nat. Med. 25, 1–11 (2019).

    Article  CAS  Google Scholar 

  21. Tabula Muris Consortium. et al. Single-cell transcriptomics of 20 mouse organs creates a Tabula Muris. Nature 562, 367–372 (2018).

    Article  CAS  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. 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 

  24. Katsimpardi, L. et al. Vascular and neurogenic rejuvenation of the aging mouse brain by young systemic factors. Science 344, 630–634 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. 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 

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

    Article  CAS  PubMed  Google Scholar 

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

    CAS  Google Scholar 

  28. Eggel, A. & Wyss-Coray, T. A revival of parabiosis in biomedical research. Swiss Med. Wkly. 144, w13914 (2014).

    PubMed  PubMed Central  Google Scholar 

  29. Lee, M.-W. et al. Activated type 2 innate lymphoid cells regulate beige fat biogenesis. Cell 160, 74–87 (2015).

    Article  CAS  PubMed  Google Scholar 

  30. Qiu, Y. et al. Eosinophils and type 2 cytokine signaling in macrophages orchestrate development of functional beige fat. Cell 157, 1292–1308 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Lee, N. A. et al. Expression of IL-5 in thymocytes/T cells leads to the development of a massive eosinophilia, extramedullary eosinophilopoiesis, and unique histopathologies. J. Immunol. 158, 1332–1344 (1997).

    CAS  PubMed  Google Scholar 

  32. Conboy, I. M., Conboy, M. J., Smythe, G. M. & Rando, T. A. Notch-mediated restoration of regenerative potential to aged muscle. Science 302, 1575–1577 (2003).

    Article  CAS  PubMed  Google Scholar 

  33. Shefer, G., Van de Mark, D. P., Richardson, J. B. & Yablonka-Reuveni, Z. Satellite-cell pool size does matter: defining the myogenic potency of aging skeletal muscle. Dev. Biol. 294, 50–66 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Fairfax, K. A. et al. Transcriptional profiling of eosinophil subsets in interleukin‐5 transgenic mice. J. Leukoc. Biol. 104, 195–204 (2018).

    Article  CAS  PubMed  Google Scholar 

  35. Ohnmacht, C., Pullner, A., van Rooijen, N. & Voehringer, D. Analysis of eosinophil turnover in vivo reveals their active recruitment to and prolonged survival in the peritoneal cavity. J. Immunol. 179, 4766–4774 (2007).

    Article  CAS  PubMed  Google Scholar 

  36. Sudo, K., Ema, H., Morita, Y. & Nakauchi, H. Age-associated characteristics of murine hematopoietic stem cells. J. Exp. Med. 192, 1273–1280 (2000).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. de Haan, G. & Van Zant, G. Dynamic changes in mouse hematopoietic stem cell numbers during aging. Blood 93, 3294–3301 (1999).

    Article  PubMed  Google Scholar 

  38. Reynaud, D. et al. IL-6 controls leukemic multipotent progenitor cell fate and contributes to chronic myelogenous leukemia development. Cancer Cell 20, 661–673 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Riether, C., Schürch, C. M. & Ochsenbein, A. F. Regulation of hematopoietic and leukemic stem cells by the immune system. Cell Death Differ. 22, 187–198 (2015).

    Article  CAS  PubMed  Google Scholar 

  40. Schürch, C. M., Riether, C. & Ochsenbein, A. F. Cytotoxic CD8+ T cells stimulate hematopoietic progenitors by promoting cytokine release from bone marrow mesenchymal stromal cells. Cell Stem Cell 14, 460–472 (2014).

    Article  PubMed  CAS  Google Scholar 

  41. Pioli, P. D., Casero, D., Montecino-Rodriguez, E., Morrison, S. L. & Dorshkind, K. Plasma cells are obligate effectors of enhanced myelopoiesis in aging bone marrow. Immunity 51, P351–P366 (2019).

  42. Akunuru, S. & Geiger, H. Aging, clonality, and rejuvenation of hematopoietic stem cells. Trends Mol. Med. 22, 701–712 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  44. Shaw, A. C., Joshi, S., Greenwood, H., Panda, A. & Lord, J. M. Aging of the innate immune system. Curr. Opin. Immunol. 22, 507–513 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Goronzy, J. J. & Weyand, C. M. Understanding immunosenescence to improve responses to vaccines. Nat. Immunol. 14, 428–436 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Salvioli, S. et al. Inflamm-aging, cytokines and aging: state of the art, new hypotheses on the role of mitochondria and new perspectives from systems biology. Curr. Pharm. Des. 12, 3161–3171 (2006).

    Article  CAS  PubMed  Google Scholar 

  47. Zwick, R. K., Guerrero-Juarez, C. F., Horsley, V. & Plikus, M. V. Anatomical, physiological, and functional diversity of adipose tissue. Cell Metab. 27, 68–83 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Zoico, E. et al. Brown and beige adipose tissue and aging. Front. Endocrinol. 10, 368 (2019).

    Article  Google Scholar 

  49. Ghosh, A. K., O’Brien, M., Mau, T., Qi, N. & Yung, R. Adipose tissue senescence and inflammation in aging is reversed by the young milieu. J. Gerontol. A 74, 1709–1715 (2019).

    Article  CAS  Google Scholar 

  50. Yu, P., Yuan, R., Yang, X. & Qi, Z. Adipose tissue, aging, and metabolism. Curr. Opin. Endocr. Metab. Res. 5, 11–20 (2019).

    Article  Google Scholar 

  51. Wu, D. et al. Aging up-regulates expression of inflammatory mediators in mouse adipose tissue. J. Immunol. 179, 4829–4839 (2007).

    Google Scholar 

  52. Franceschi, C., Garagnani, P., Vitale, G., Capri, M. & Salvioli, S. Inflammaging and ‘Garb-aging’. Trends Endocrinol. Metab. 28, 199–212 (2017).

    Article  CAS  PubMed  Google Scholar 

  53. Kusminski, C. M., Bickel, P. E. & Scherer, P. E. Targeting adipose tissue in the treatment of obesity-associated diabetes. Nat. Rev. Drug Discov. 15, 639–660 (2016).

    Article  CAS  PubMed  Google Scholar 

  54. Starr, M. E., Saito, M., Evers, B. M. & Saito, H. Age-associated increase in cytokine production during systemic inflammation—II: the role of IL-1β in age-dependent IL-6 upregulation in adipose tissue. J. Gerontol. A 70, 1508–1515 (2015).

    Article  CAS  Google Scholar 

  55. Soysal, P. et al. Inflammation and frailty in the elderly: a systematic review and meta-analysis. Ageing Res. Rev. 31, 1–8 (2016).

    Article  CAS  PubMed  Google Scholar 

  56. Oh, J., Lee, Y. D. & Wagers, A. J. Stem cell aging: mechanisms, regulators and therapeutic opportunities. Nat. Med. 20, 870–880 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Geiger, H. & Van Zant, G. The aging of lympho-hematopoietic stem cells. Nat. Immunol. 3, 329–333 (2002).

    Article  CAS  PubMed  Google Scholar 

  58. Vogelaar, C. F. et al. Fast direct neuronal signaling via the IL-4 receptor as therapeutic target in neuroinflammation. Sci. Transl. Med. 10, eaao2304 (2018).

    Article  PubMed  CAS  Google Scholar 

  59. Schneider, C. A., Rasband, W. S. & Eliceiri, K. W. NIH Image to ImageJ: 25 years of image analysis. Nat. Methods 9, 671–675 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Liu, L., Cheung, T. H., Charville, G. W. & Rando, T. A. Isolation of skeletal muscle stem cells by fluorescence-activated cell sorting. Nat. Protoc. 10, 1612–1624 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Dyer, K. D. et al. Functionally competent eosinophils differentiated ex vivo in high purity from normal mouse bone marrow. J. Immunol. 181, 4004–4009 (2008).

    Article  CAS  PubMed  Google Scholar 

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

    Google Scholar 

  63. R Core Team. R: A Language and Environment for Statistical Computing (R Foundation for Statistical Computing, 2014).

  64. van den Berg, R. A., Hoefsloot, H. C. J., Westerhuis, J. A., Smilde, A. K. & van der Werf, M. J. Centering, scaling, and transformations: improving the biological information content of metabolomics data. BMC Genomics 7, 142 (2006).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  65. de Hoon, M. J. L., Imoto, S., Nolan, J. & Miyano, S. Open source clustering software. Bioinformatics 20, 1453–1454 (2004).

    Article  CAS  PubMed  Google Scholar 

  66. Saldanha, A. J. Java Treeview—extensible visualization of microarray data. Bioinformatics 20, 3246–3248 (2004).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We thank past and present members of the Noti, Eggel and Wyss-Coray laboratories for discussions and valuable input; A.-L. Huguenin, U. Lüthi and C. Krüger for technical support; J. J. Lee and H.-U. Simon for providing IL-5 transgenic mice; M. Fux, A. Odermatt and the DBMR FACSlab for technical support with cell sorting; the staff and animal caretakers of the Central Animal Facility in Bern for their advice and support; K. Rufibach for valuable discussions; and H. Maeker and the Stanford Human Immune Monitoring Center for their help with qPCR multiplex array. J.A. is supported by grants from École Polytechnique Fédérale de Lausanne, the Swiss National Science Foundation (no. 31003A-140780), the AgingX programme of the Swiss Initiative for Systems Biology (no. 51RTP0-151019) and the NIH (no. R01AG043930). This work was funded by the National Institute on Aging (no. R01AG053382 to S.A.V. and nos. AG045034 and DP1AG053015 to T.W.-C.), the Department of Veterans Affairs (to T.W.-C.), the Glenn Center for the Biology of Aging (to T.W.-C.), a Novartis grant for Medical-Biological Research (no. 15B100 to M.N.), a FreeNovation Novartis grant (to M.N) and by grants from the Fondation Acteria (to A.E. and M.N.) and Velux Stiftung (no. 1095 to A.E.).

Author information

Authors and Affiliations

Authors

Contributions

M.N. and A.E. designed the project. D.B., C.R., R.v.B., K.I.M., A.S., Z.D., N.Z., P. Gasser, P. Guntern, H.Y., J.M.C., M.B., M.H., D.G., M.S., N.M., S.A.V., M.N. and A.E. performed the experiments. F.S. and N.G.-R. provided human samples. W.H., S.L.L., P.M.V., J.A., S.A.V. and T.W.-C. provided important advice. D.B., M.N. and A.E. analysed the data and wrote the manuscript.

Corresponding authors

Correspondence to Mario Noti or Alexander Eggel.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Peer review information Primary Handling Editor: Pooja Jha.

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

Extended data

Extended Data Fig. 1 Tissue screens for CCL11 and CCL2 protein expression in young and aged mice.

a, Comparison of CCL11 and CCL2 protein expression in muscle, spleen, brain, kidney, thymus, liver, lung, skin, mesenteric lymph nodes (MLN), colon, subcutaneous WAT (scWAT) and epidydimal WAT (eWAT) of young (Y, 2-3 months) and aged mice (A, 18-20 months) as assessed by western blot. Tissues of three biologically independent animals were pooled. HSP90 served as loading control. Quantification of b, CCL11 and c, CCL2 protein levels normalized to HSP90 in indicated tissues of young (Y, 2-3 months) and aged mice (A, 18-20 months) by ImageJ. One out of two independently performed experiments is shown. d, Comparison of CCL11 and CCL2 mRNA expression levels in indicated tissues of aged mice (18-20 months) as assessed by qPCR (n=4). e, CCL2 and CCL11 protein levels were assessed by western blot (n=4). One out of 3 independently performed experiments is shown. f, Quantification of CCL11 and CCL2 protein levels normalized to HSP90 in indicated tissues of young (Y, 2-3 months, n=4) and aged mice (A, 18-20 months, n=4). HSP90 served as loading control. Protein levels from total eWAT was calculated. Statistical significance was calculated by one-way ANOVA followed by two-tailed post-hoc Dunnett’s multiple comparison test against eWAT (d) or by unpaired two-tailed Student’s t test between young and aged samples (f). Data are shown as individual data points with mean ± SEM. *p < 0.05, **p < 0.01, ***p < 0.001. ns, not significant. Uncropped western blots are provided in the Source Data File.

Source data

Extended Data Fig. 2 Gating strategies for human and mouse ATEs and age-related adipose tissue hypertrophy.

a, Gating strategy for human omental adipose tissue eosinophils. b, Representative images of H&E stained human omental adipose tissue from young and aged donors of indicated age. Scale bar: 100 μm. c, Quantification of adipocyte size in H&E stained sections from human omental adipose tissue (n=17 biologically independent human donors) by ImageJ. d, Representative gating strategy for ATE (F4/80int, SiglecF+) and ATM (F4/80+, SiglecF-) in mouse visceral adipose tissue of young (3 months) and aged mice (20 months). e, Absolute eosinophil and macrophage numbers per gram eWAT of young (3 months, n=10) and aged mice (20 months, n=10). This experiment was done once. f, Representative photographs of H&E stained histological eWAT sections of indicated treatment groups. g, Quantification of adipocyte hypertrophy in young (n=20), Aged-PBS (n=19), Aged-yEOS (n=22) and Aged-yEOSIL-4-/- (n=20) biologically independent mice by ImageJ. Statistical significance was calculated by unpaired two-tailed Student’s t test (e), the Pearson correlation coefficient between adipocyte size and age (c) or by one-way ANOVA followed by two-tailed post-hoc Dunnett’s multiple comparison test against the aged-PBS treated group (g). Data are shown as individual data points with mean bars ± SEM. *p < 0.05, **p < 0.01, ***p < 0.001. ns, not significant.

Extended Data Fig. 3 Recruitment of sort-purified GFP+ eosinophils to WAT of aged mice.

Aged mice (18 months) were adoptively transferred with sort-purified GFP+ eosinophils derived from IL-5 transgenic mice on two subsequent days. The following day eosinophil recruitment into different tissues was assessed by flow cytometry. a, Representative flow plots of transferred GFP+ and endogenous GFP tissue eosinophils in indicated tissues. b, Frequencies of transferred GFP+ and endogenous GFP tissue eosinophils in indicated tissues. c, Siglec-F surface expression on adipose tissue eosinophils from Young, Aged-PBS and Aged-yEOS -treated mice as assessed by flow cytometry. d, Representative histograms of Siclec-F expression on ATEs of indicated groups. e, Quantification of Siglec-F surface expression (MFI) on adipose tissue eosinophils of Young (n=5), Aged-PBS (n=5) and Aged-yEOS (n=4) mice. The experiment was done once. Statistical significance was calculated by one-way ANOVA followed by two-tailed post-hoc Dunnett’s multiple comparison test against the young group. ns, not significant. Data are representative of n=4 per group and are shown as mean ± SEM.

Extended Data Fig. 4 Transfer of neutrophils to aged mice does not alter WAT inflammation, hematopoetic stem cell pol or physical performance.

a, Experimental protocol. b, Frequencies of eosinophils, macrophages and calculated eosinophil:macrophage ratios in eWAT of Aged-PBS (n=8) and Aged-yNEU (n=9) mice. c, mRNA expression levels for Tnfα, Il1β and IL6 in eWAT of Aged-PBS (n=8) and Aged-yNEU mice (n=9). Data are presented as fold induction over Aged-PBS controls. d, Average time on Rotarod of Aged-PBS controls (n=8) and Aged-yNEU mice (n=9). e, Total numbers of lin, Sca-1+, c-kit+ hematopoietic stem cells (LSKs) in Aged-PBS (n=4) and Aged-yNEU (n=4) mice. f, Numbers of common myeloid progenitors (CMP), common lymphoid progenitors (CLP), and granulocyte/monocyte progenitors (GMP) in the bone marrow of Aged-PBS (n=4) and Aged-yNEU (n=4) mice. g, Frequencies of neutrophils in eWAT of young (n=5), Aged-PBS (n=4) and Aged-yNeu (n=5) mice. h, Experimental protocol of bone marrow derived eosinophil (BMDE) transfers. i, Calculated ATE:ATM ratios in eWAT of Aged-PBS (n=5) and Aged-BMDE (n=4) mice as measured by flow cytometry. j, IL-6 protein levels in eWAT of Aged-PBS (n=6) and Aged-BMDE (n=5) mice. k, Pre- and post-treatment IL-6 plasma protein levels in Aged-PBS (n=6) and Aged-BMDE (n=5). l, Intra-group and m, inter-group comparison of pre- and post-treatment average time on wheel (Rotarod test) in Aged-PBS (n=6) and Aged-BMDE (n=5) mice. Delta in performances in (l) is calculated relative to baseline (post- minus pre-treatment results). Statistical significance was calculated by Wilcoxon matched pairs signed rank test (k, l), by unpaired two-tailed Student’s t test (b, c, d, e, f, I, j, m) or by one-way ANOVA followed by two-tailed post-hoc Dunnett’s multiple comparison test against the aged-PBS treated group (g). Data are pooled from two independently performed experiments (except for (g-m) only one experiment has been performed) and shown as individual data points with mean ± SEM. Data are shown as mean ± SEM. *p < 0.05, **p < 0.01. ns, not significant.

Extended Data Fig. 5 Eosinophil transfers do not alter age-related changes in murine subcutaneous WAT.

a, Gating strategy for ATMs and ATEs in scWAT of young (3 months) and aged (20 months) mice. b, Calculated ATE:ATM ratio in scWAT of young (n=10), Aged-PBS (n=10), Aged-yEOS (n=6) and Aged-yEOSIL-4-/- (n=7) mice. c, Representative photographs of H&E stained histological scWAT sections of indicated treatment groups. d, Quantification of adipocyte hypertrophy in Young (n=11), Aged-PBS (n=10), Aged-yEOS (n=9) and Aged-yEOSIL-4-/- (n=12) mice by ImageJ. e, IL-6 and CCL2 protein levels in scWAT of Young (n=15), Aged-PBS (n=14), Aged-yEOS (n=12) and Aged-yEOSIL-4-/- (n=13) mice. Statistical significance was calculated by one-way ANOVA followed by two-tailed post-hoc Dunnett’s multiple comparison test against the aged-PBS treated group. Data (e) are pooled from 2 independently performed experiments or performed once (a-d) and shown as individual data points with mean bars ± SEM. *p < 0.05, **p < 0.01, ***p < 0.001. ns, not significant.

Extended Data Fig. 6 Glucose metabolism in response to eosinophil transfers to aged mice.

a, Blood glucose levels in Aged-PBS (20 months, n=4) and Aged-yEOS (n=5) mice in response to i.p. glucose challenge over time. b, Calculated HOMA-IR for Aged-PBS (20 months, n=4) and Aged-yEOS (20 months, n=5) mice. Statistical significance was tested by unpaired two-tailed Student’s t-test. One out of 3 independently performed experiments is shown. Data are shown as individual data points with mean bars ± SEM. ns, not significant.

Extended Data Fig. 7 Open field activity tests.

a, Mean velocity, total distance, accumulative mobility- and immobility of Aged-PBS, Aged-yEOS and Aged-yNEU mice (n=4 per group). b, Representative heat maps for Aged-PBS, Aged-yEOS and Aged-yNEU mice demonstrating the animal’s position in the arena. The experiment was done once. Statistical significance was calculated by one-way ANOVA followed by two-tailed post-hoc Dunnett’s multiple comparison test against the aged-PBS treated group. Data are shown as mean bars ± SEM. ns, not significant.

Extended Data Fig. 8 Transfer of young eosinophils is associated with alterations in muscle stem cell frequencies but not function.

a, Gating strategy and representative flow plots of CD31, CD45, Sca-1, Vcam+ and integrin α7+ satellite cells in muscle of Young (n=13), Aged-PBS (n=17), Aged-yEOS, (n=17) and Aged-yEOSIL-4-/- (n=17) mice. b, Quantification of muscle stem cell frequencies in indicated groups. c, Representative photographs of immunofluorescent stained sort-purified and differentiated satellite cells. d, Quantification of cell colony formation of sort-purified muscle stem cells of Young (n=10), Aged-PBS (n=10), Aged-yEOS (n=8) and Aged-yEOSIL-4-/- (n=8) mice e, Representative H&E stained longitudinal and cross-sectional quadriceps femoris in indicated groups. f, Quantification of centrally nucleated myofibers in sections of Young (n=26), Aged-PBS (n=26), Aged-yEOS (n=18) and Aged-yEOSIL-4-/- (n=13) mice. g, Muscle weight (femur) was measured in Young (n=5), Aged-PBS (n=9), Aged-yEOS (n=8) and Aged-yEOSIL-4-/- (n=7) mice. Data (a-f) are pooled from 2 independently performed experiments except for g (one experiment has been performed). Statistical significance was calculated by one-way ANOVA followed by two-tailed post-hoc Dunnett’s multiple comparison test against the aged-PBS treated group. Data are shown as individual data points with mean ± SEM. *p < 0.05, **p < 0.01, ***p < 0.001. ns, not significant.

Extended Data Fig. 9 Eosinophil transfers reverse myeloid skewing in old age.

a, Gating strategy for the identification of LSK, HSC-SLAM, HSC-AD, CMP, CLP and GMP populations. b, Absolute numbers of CMP, CLP and GMP per mouse in Young (n=5), Aged-PBS (n=7) and Aged-yEOS (n=8) groups. c, Absolute numbers of CMP, CLP and GMP per mouse in Aged-PBS (n=8) and Aged-yEOSIL-4-/- (n=10) groups. One out of two independently performed experiments is shown. Statistical significance in (b) was calculated one-way ANOVA followed by two-tailed post-hoc Dunnett’s multiple comparison test against the aged-PBS treated group and (c) by two-tailed Student’s t-test. d, Gating strategy for the identification of germinal GCB. Data are shown as individual data points with mean ± SEM. *p < 0.05, **p < 0.01, ***p < 0.001.

Extended Data Fig. 10 Eosinophils adopt a senescent-like inflammatory phenotype with age.

a, Heat map representing Ct values of p21, Vegfa, Il1b, Il6 and Tnfa in sort-purified, blood-derived eosinophils from aged WT (20 months), young WT (3 months) or young IL5 transgenic mice (3 months) as assessed by targeted fluidigm qPCR array. b, Relative expression levels of Tnfa, Il1b, Il6, p21 and Vegfa in sort-purified, blood-derived eosinophils from aged WT (20 months), young WT (3 months) or young IL5 transgenic mice (3 months) as assessed by fluidigm qPCR array. Eosinophils from 3 animals were pooled for each measurement (n=3 per group). The experiment was done once. c, Relative expression levels of TNFa, IL1b, IL6, p21 and VEGFA in human blood derived eosinophils from young (average age=34, n=8) and aged (average age=64, n=7) donors. Data in (b and c) are shown as individual data points with mean ± SEM and statistical significance was calculated by one-way ANOVA followed by two-tailed post-hoc Dunnett’s multiple comparison test against the young group (b) and two-tailed students t-test (c). *p < 0.05, **p < 0.01, ***p < 0.001. ns, not significant. ND, not detectable.

Supplementary information

Source data

Source Data Extended Data Fig. 1.

Unprocessed immunolots.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Brigger, D., Riether, C., van Brummelen, R. et al. Eosinophils regulate adipose tissue inflammation and sustain physical and immunological fitness in old age. Nat Metab 2, 688–702 (2020). https://doi.org/10.1038/s42255-020-0228-3

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s42255-020-0228-3

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing