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.

  • Letter
  • Published:

Induction of mitochondrial recycling reverts age-associated decline of the hematopoietic and immune systems

Subjects

Abstract

Aging compromises hematopoietic and immune system functions, making older adults especially susceptible to hematopoietic failure, infections and tumor development, and thus representing an important medical target for a broad range of diseases. During aging, hematopoietic stem cells (HSCs) lose their blood reconstitution capability and commit preferentially toward the myeloid lineage (myeloid bias)1,2. These processes are accompanied by an aberrant accumulation of mitochondria in HSCs3. The administration of the mitochondrial modulator urolithin A corrects mitochondrial function in HSCs and completely restores the blood reconstitution capability of ‘old’ HSCs. Moreover, urolithin A-supplemented food restores lymphoid compartments, boosts HSC function and improves the immune response against viral infection in old mice. Altogether our results demonstrate that boosting mitochondrial recycling reverts the aging phenotype in the hematopoietic and immune systems.

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: UA in vitro treatment reverses age-associated defects in HSCs.
Fig. 2: UA oral supplementation rejuvenates the hematopoietic system of old mice.
Fig. 3: UA supplementation improves the CD8+ T cell response to viral infection in old mice.
Fig. 4: UA induces mitochondrial recycling and restores mitochondrial fitness.

Similar content being viewed by others

Data availability

All data and materials are available from the corresponding author to any researcher for the purpose of reproducing or extending the analyses. Source data are provided with this paper.

References

  1. Morrison, S. J., Wandycz, A. M., Akashi, K., Globerson, A. & Weissman, I. L. The aging of hematopoietic stem cells. Nat. Med. 2, 1011–1016 (1996).

    Article  CAS  PubMed  Google Scholar 

  2. Pang, W. W. et al. Human bone marrow hematopoietic stem cells are increased in frequency and myeloid-biased with age. Proc. Natl Acad. Sci. USA 108, 20012–20017 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Ho, T. T. et al. Autophagy maintains the metabolism and function of young and old stem cells. Nature 543, 205–210 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. 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 

  5. Chandel, N. S., Jasper, H., Ho, T. T. & Passegué, E. Metabolic regulation of stem cell function in tissue homeostasis and organismal ageing. Nat. Cell Biol. 18, 823–832 (2016).

    Article  CAS  PubMed  Google Scholar 

  6. Hine, C. & Mitchell, J. R. Saying no to drugs: fasting protects hematopoietic stem cells from chemotherapy and aging. Cell Stem Cell 14, 704–705 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Flach, J. et al. Replication stress is a potent driver of functional decline in ageing haematopoietic stem cells. Nature 512, 198–202 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Beerman, I., Seita, J., Inlay, M. A., Weissman, I. L. & Rossi, D. J. Quiescent hematopoietic stem cells accumulate DNA damage during aging that is repaired upon entry into cell cycle. Cell Stem Cell 15, 37–50 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Beerman, I. & Rossi, D. J. Epigenetic regulation of hematopoietic stem cell aging. Exp. Cell Res. 329, 192–199 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Chambers, S. M. et al. Aging hematopoietic stem cells decline in function and exhibit epigenetic dysregulation. PLoS Biol. 5, e201 (2007).

    Article  PubMed  PubMed Central  Google Scholar 

  11. Sahin, E. & Depinho, R. A. Linking functional decline of telomeres, mitochondria and stem cells during ageing. Nature 464, 520–528 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Nakada, D., Saunders, T. L. & Morrison, S. J. Lkb1 regulates cell cycle and energy metabolism in haematopoietic stem cells. Nature 468, 653–658 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Gurumurthy, S. et al. The Lkb1 metabolic sensor maintains haematopoietic stem cell survival. Nature 468, 659–663 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Gan, B. et al. Lkb1 regulates quiescence and metabolic homeostasis of haematopoietic stem cells. Nature 468, 701–704 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Zhang, J. et al. UCP2 regulates energy metabolism and differentiation potential of human pluripotent stem cells. EMBO J. 30, 4860–4873 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Suda, T., Takubo, K. & Semenza, G. L. Metabolic regulation of hematopoietic stem cells in the hypoxic niche. Cell Stem Cell 9, 298–310 (2011).

    Article  CAS  PubMed  Google Scholar 

  17. Ito, K. et al. Self-renewal of a purified Tie2+ hematopoietic stem cell population relies on mitochondrial clearance. Science 354, 1156–1160 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Vannini, N. et al. The NAD-booster nicotinamide riboside potently stimulates hematopoiesis through increased mitochondrial clearance. Cell Stem Cell 24, 405–418 (2019).

    Article  CAS  PubMed  Google Scholar 

  19. Mohrin, M. et al. Stem cell aging. A mitochondrial UPR-mediated metabolic checkpoint regulates hematopoietic stem cell aging. Science 347, 1374–1377 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Cheng, C.-W. et al. Prolonged fasting reduces IGF-1/PKA to promote hematopoietic-stem-cell-based regeneration and reverse immunosuppression. Cell Stem Cell 14, 810–823 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Sun, X. et al. Nicotinamide riboside attenuates age-associated metabolic and functional changes in hematopoietic stem cells. Nat. Commun. 12, 2665 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Larrosa, M., García-Conesa, M. T., Espín, J. C. & Tomás-Barberán, F. A. Ellagitannins, ellagic acid and vascular health. Mol. Aspects Med. 31, 513–539 (2010).

    Article  CAS  PubMed  Google Scholar 

  24. Ryu, D. et al. Urolithin A induces mitophagy and prolongs lifespan in C. elegans and increases muscle function in rodents. Nat. Med. 22, 879–888 (2016).

    Article  CAS  PubMed  Google Scholar 

  25. Luan, P. et al. Urolithin A improves muscle function by inducing mitophagy in muscular dystrophy. Sci. Transl. Med. 13, eabb0319 (2021).

    Article  CAS  PubMed  Google Scholar 

  26. Andreux, P. A. et al. The mitophagy activator urolithin A is safe and induces a molecular signature of improved mitochondrial and cellular health in humans. Nat. Metab. 1, 595–603 (2019).

    Article  CAS  PubMed  Google Scholar 

  27. D’Amico, D. et al. Impact of the natural compound urolithin A on health, disease, and aging. Trends Mol. Med. 27, 687–699 (2021).

    Article  PubMed  Google Scholar 

  28. Beerman, I. et al. Functionally distinct hematopoietic stem cells modulate hematopoietic lineage potential during aging by a mechanism of clonal expansion. Proc. Natl Acad. Sci. USA 107, 5465–5470 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Ho, Y. H. et al. Remodeling of bone marrow hematopoietic stem cell niches promotes myeloid cell expansion during premature or physiological aging. Cell Stem Cell 25, 407–418 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Decman, V. et al. Defective CD8 T cell responses in aged mice are due to quantitative and qualitative changes in virus-specific precursors. J. Immunol. 188, 1933–1941 (2012).

    Article  CAS  PubMed  Google Scholar 

  31. Ucar, D. et al. The chromatin accessibility signature of human immune aging stems from CD8+ T cells. J. Exp. Med. 214, 3123–3144 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. McWilliams, T. G. et al. mito-QC illuminates mitophagy and mitochondrial architecture in vivo. J. Cell Biol. 214, 333–345 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Klionsky, D. J. et al. Guidelines for the use and interpretation of assays for monitoring autophagy (4th edition)1. Autophagy 17, 1–382 (2021).

    Article  PubMed  PubMed Central  Google Scholar 

  34. Mansell, E. et al. Mitochondrial potentiation ameliorates age-related heterogeneity in hematopoietic stem cell function. Cell Stem Cell 28, 241–256 (2021).

    Article  CAS  PubMed  Google Scholar 

  35. Jovaisaite, V., Mouchiroud, L. & Auwerx, J. The mitochondrial unfolded protein response, a conserved stress response pathway with implications in health and disease. J. Exp. Biol. 217, 137–143 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Battegay, M. et al. Quantification of lymphocytic choriomeningitis virus with an immunological focus assay in 24- or 96-well plates. J. Virol. Methods 33, 191–198 (1991).

    Article  CAS  PubMed  Google Scholar 

  37. Vannini, N. et al. Specification of haematopoietic stem cell fate via modulation of mitochondrial activity. Nat. Commun. 7, 13125 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank G. Coukos (LICR, University of Lausanne (UNIL), Centre Hospitalier Universitaire Vaudois) for critical feedback and F. Derouet (UNIL) for animal care. Flow cytometry analysis and sorting was performed at the Flow Cytometry Facility at UNIL with the help of R. Bedel, F. Sala de Oyanguren, K. Blackney and A. Wilson. We thank Nestlé Health Science for their suggestions about the study. M.G. was partially funded by a collaborative Jebsen Foundation grant (EPFL/UNIL) to N.V. and O.N. The N.V. laboratory is supported by the Swiss Cancer Research Foundation (KFS-4993-02-2020-R) and San Salvatore Foundation. The J.A. laboratory is supported by grants from EPFL, the European Research Council (ERC-AdG-787702), the Swiss National Science Foundation (SNSF) (31003A_179435) and a Global Research Laboratory grant of the National Research Foundation of Korea (2017K1A1A2013124). F.S. is supported by the SNSF (323530_183986). The O.N. laboratory was supported by SNSF grant nos. PP00P3_183725 and CRSII5-186271. P.-C.H. is funded in part by the European Research Council Staring Grant (802773-MitoGuide), an SNSF project grant (no. 31003A_182470), the Cancer Research Institute (Lloyd J. Old STAR Award) and LICR. W.H. is funded in part by a grant from the SNSF (no. 310030_200898). All 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

M.G., Y-H.C. and N.V. conceived the ideas, designed the experiments and analyzed the results. M.G., Y-H.C., N.V. and F.S. performed the HSC experiments. M.C., H.C.H. and W.H. performed and interpreted the infection model experiments. P.G. evaluated the impact of UA dietary supplementation on secondary lymphoid tissues and performed and interpreted the cell respiration analyses. J.A. and O.N. conceived the ideas and provided key reagents and samples. S.C. provided the human samples for HSC purification. O.N. and C.B. performed the human HSC experiments. F.F., Y-R.Y., H.G. and P.-C.H. provided the animal model and helped with data interpretation. N.V. wrote the manuscript. All authors edited and reviewed the final manuscript.

Corresponding author

Correspondence to Nicola Vannini.

Ethics declarations

Competing interests

J.A. is a scientific advisor to Amazentis, a company that develops UA as a therapeutic agent. Some elements of this work are part of patent no. 19152437.0 of which N.V. and M.G. are inventors. The remaining authors declare no competing interests.

Peer review

Peer review information

Nature Aging thanks Saghi Ghaffari 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 Old HSCs have higher mitochondria content.

Mitochondrial DNA (mtDNA) quantification by QPCR expressed as fold change compared to young HSCs. Young values are normalized on their mean value (P-value = 0.0571) (n = 4; values are mean ± s.e.m.; two-tailed Mann-Whitney test; * P ≤ 0.05, ** P ≤ 0.01, *** P ≤ 0.001).

Source data

Extended Data Fig. 2 Donor chimerism analysis in the bone marrow.

a, Lymphoid vs myeloid compartments of primary transplants in peripheral blood at the indicated timepoints. b, Analysis of donor derived contribution for lymphoid and myeloid lineages in bone marrow at the endpoint of primary transplant (Lymphoid: young ctrl vs old ctrl P-value = 0.0003; old ctrl vs old UA P-value = 0.0011; myeloid: young ctrl vs old ctrl P-value = 0.0002; old ctrl vs old UA P-value = 0.0006;). c, Analysis of donor derived HSCs at the endpoint of primary transplant. d, Analysis of donor derived HSCs at the endpoint of secondary transplant (n = 10; values are mean ± s.e.m.; two-tailed Student’s t test; * P ≤ 0.05, ** P ≤ 0.01, *** P ≤ 0.001, **** P ≤ 0.0001). e, HSCs isolated from bone marrow of young mice were cultured for three days in the presence of 20μM UA and transplanted in lethally irradiated primary recipient together with bone marrow derived from competitor mice. Blood donor chimerism analyses of primary transplant at indicated timepoints (n = 10; values are mean ± s.e.m.).

Source data

Extended Data Fig. 3 UA improves Colony forming capacity (CFU assay) of old human HSCs.

a, 1000 CD34+CD38CD45RA- cells derived from the bone marrow of anonymous old adults (58-77 years old) were treated for 3 days with 20μM UA, and colony formations was measured at 15 days post seeding and replated for secondary CFUs. b, analysis of primary CFUs (P-value < 0.0001) (n = 14; values are mean ± s.e.m.; two-tailed Student’s t test; * P ≤ 0.05, ** P ≤ 0.01, *** P ≤ 0.001). c, Assessment of long-term hematopoietic capacity by secondary CFUs analysis (P-value = 0.0042) (n = 8; values are mean ± s.e.m.; two-tailed Student’s t test; * P ≤ 0.05, ** P ≤ 0.01, *** P ≤ 0.001).

Source data

Extended Data Fig. 4 Peripheral blood analyses of UA fed mice.

Peripheral blood cell analyses of old mice fed with ctrl (old ctrl) or UA enriched diet (old UA). Analyses were performed at the indicated time points with blood cell counter and normalized to day 0 (WBC: white blood cell; Lym: Lymphocytes; Gra: Granulocytes; Mon: Monocytes; RBC: red blood cells; HGB: hemoglobin) (n = 8; values are mean ± s.e.m.;).

Source data

Extended Data Fig. 5 Hematopoietic stem and progenitor analyses of old mice supplemented with UA.

a, Gating strategy to identify hematopoietic stem and progenitor populations from the BM for endpoint analysis. HSCs: LineagecKit+Sca1+ (LKS) CD150+CD48; Multipotent progenitors (MPPs): LKS CD150, Common lymphoid progenitor (CLP): CD127+ cKitlow Sca1low; Committed progenitors (cKit+) include common myeloid progenitors (CMPs: LineagecKit+Sca1(KLS) FcRlowCD34+), granulocyte–macrophage progenitors (GMPs: KLS-FcRlowCD34 + ), and megakaryocyte-erythroid progenitors (MEPs: KLSFcRCD34). b, Analyses of lymphoid vs myeloid compartments of primary transplants at the indicated timepoints (n = 10; values are mean ± s.e.m). c, Donor chimerism analysis of secondary recipient mice in peripheral blood (Total: 4 weeks P-value = 0.0013; 8 weeks P-value < 0.0001; 12 weeks P-value = 0.0003; Lymphoid: 4 weeks P-value < 0.0001; 8 weeks P-value < 0.0001; 12 weeks P-value = 0.0002; Myeloid: 4 weeks P-value = 0.0089; 8 weeks P-value = 0.0049; 12 weeks P-value = 0.0153) (n = 5; values are mean ± s.e.m.; two-tailed Student’s t test; * P ≤ 0.05, ** P ≤ 0.01, *** P ≤ 0.001).

Source data

Extended Data Fig. 6 Expression of inhibitory receptors markers and cytokines by virus-specific CD8 + T cells.

a, Expression of IFNγ (gp33 + : young ctrl vs old UA P-value = 0.0112; np396+: young ctrl vs old UA P-value = 0.0034; old ctrl vs old UA P-value = 0.0160), TNFα (gp33 + :young ctrl vs old ctrl P-value < 0.0001; young ctrl vs old UA P-value = 0.0003; np396+:young ctrl vs old ctrl P-value < 0.0001; young ctrl vs old UA P-value < 0.0001) and IL-2 by CD8+ T cells in response to restimulation with gp33 or np396 peptides. b, Expression of co- inhibitory receptors PD-1 (gp33 + :young ctrl vs old ctrl P-value < 0.0001; young ctrl vs old UA P-value = 0.0125; old ctrl vs old UA P-value = 0.0146 np396+:young ctrl vs old ctrl P-value < 0.0001; young ctrl vs old UA P-value = 0.0025; old ctrl vs old UA P-value = 0.0159)and Lag3 (gp33 + :young ctrl vs old ctrl P-value < 0.0001; young ctrl vs old UA P-value = 0.0007; old ctrl vs old UA P-value = 0.0173; np396+:young ctrl vs old ctrl P-value < 0.0001; young ctrl vs old UA P-value < 0.0001; old ctrl vs old UA P-value = 0.0301) by gp33+ and np396 CD44+ CD8+ T cells (n = 10; values are mean ± s.e.m.; two-way ANOVA; * P ≤ 0.05, ** P ≤ 0.01, *** P ≤ 0.001, **** P ≤ 0.0001).

Source data

Extended Data Fig. 7 Viral response of CD8 + T cells derived from old UA treated HSCs.

a, Quantification of donor derived (CD45.1+) virus specific (gp33+ or np396+) CD8+ T cells (gp33 + :young ctrl vs old ctrl P-value = 0.0407; np396+:young ctrl vs old ctrl P-value = 0.0298). b, Proportion of activated KLRG1+ within donor derived virus specific CD8+ T. (n = 5; values are mean ± s.e.m.; two-way ANOVA; * P ≤ 0.05, ** P ≤ 0.01, *** P ≤ 0.001, **** P ≤ 0.0001).

Source data

Extended Data Fig. 8 UA in vitro treatment modulates T cell function and mitochondrial profiles.

a, Splenocytes derived from old mice were activated at day0 and restimulated at day5 with CD3/CD28 beads cultured with 5 μM UA. b, Analyses of mitochondrial activity (TMRM) and mass (MTG, mitotracker Green) in CD8+ and CD4 + T cells measured as mean fluorescence intensity (MFI). c, Analyses of the proportion of cells INFγ+TNFα+ upon restimulation with CD3/CD28 beads at day 5 (n = 4; values are mean ± s.e.m.; two-tailed paired Student’s t test; * P ≤ 0.05, ** P ≤ 0.01, *** P ≤ 0.001).

Source data

Extended Data Fig. 9 UA modulates mitochondrial recycling in HSCs.

a, Analysis of mitophagy induction in HSCs purified from mito-QC mice measured as MFI ratio between GFP and mCherry signals(P-value = 0.0037). (n = 4; two tailed paired Student’s t test; * P ≤ 0.05, ** P ≤ 0.01, *** P ≤ 0.001). b, Expression of CD150 measured by mean fluorescence intensity (MFI) in young and old HSCs (P-value = 0.0009)(n = 4; values are mean ± s.e.m.; two-tailed Student’s t test; * P ≤ 0.05, ** P ≤ 0.01, *** P ≤ 0.001). c, HSCs derived from Mx1-PARK2 mice (CD45.2) were transplanted in lethally irradiated recipient mice (CD45.1/2) together with total bone marrow competitor cells (CD45.1).d, Blood donor chimerism of primary transplant at the indicated timepoint. e, Lymphoid vs myeloid compartments of primary transplants at the indicated timepoints (Ctrl vs PARK2-/- P-value = 0.0002; Ctrl vs PARK2-/- UA P-value = 0.0006) (n = 10; values are mean ± s.e.m.; two-way ANOVA; * P ≤ 0.05, ** P ≤ 0.01, *** P ≤ 0.001).

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 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.

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

Girotra, M., Chiang, YH., Charmoy, M. et al. Induction of mitochondrial recycling reverts age-associated decline of the hematopoietic and immune systems. Nat Aging 3, 1057–1066 (2023). https://doi.org/10.1038/s43587-023-00473-3

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s43587-023-00473-3

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