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.

  • Review Article
  • Published:

Metabolic regulation of somatic stem cells in vivo

Abstract

Metabolism has been studied mainly in cultured cells or at the level of whole tissues or whole organisms in vivo. Consequently, our understanding of metabolic heterogeneity among cells within tissues is limited, particularly when it comes to rare cells with biologically distinct properties, such as stem cells. Stem cell function, tissue regeneration and cancer suppression are all metabolically regulated, although it is not yet clear whether there are metabolic mechanisms unique to stem cells that regulate their activity and function. Recent work has, however, provided evidence that stem cells do have a metabolic signature that is distinct from that of restricted progenitors and that metabolic changes influence tissue homeostasis and regeneration. Stem cell maintenance throughout life in many tissues depends upon minimizing anabolic pathway activation and cell division. Consequently, stem cell activation by tissue injury is associated with changes in mitochondrial function, lysosome activity and lipid metabolism, potentially at the cost of eroding self-renewal potential. Stem cell metabolism is also regulated by the environment: stem cells metabolically interact with other cells in their niches and are able to sense and adapt to dietary changes. The accelerating understanding of stem cell metabolism is revealing new aspects of tissue homeostasis with the potential to promote tissue regeneration and cancer suppression.

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: Approaches for the analysis of stem cell metabolism: metabolomics and isotope tracing.
Fig. 2: Metabolic regulation of quiescent and activated haematopoietic stem cells.
Fig. 3: Autophagy and lysosome function in stem cells enforce quiescence and promotes the maintenance of self-renewal potential.

Similar content being viewed by others

References

  1. Agathocleous, M. et al. Ascorbate regulates haematopoietic stem cell function and leukaemogenesis. Nature 549, 476–481 (2017). This work shows that the metabolic profile of HSCs is distinct from that of restricted progenitors and that ascorbate is required to regulate HSC function, haematopoietic regeneration and leukaemia suppression.

    Article  PubMed  PubMed Central  Google Scholar 

  2. Rodriguez-Colman, M. J. et al. Interplay between metabolic identities in the intestinal crypt supports stem cell function. Nature 543, 424–427 (2017).

    Article  CAS  PubMed  Google Scholar 

  3. DeVilbiss, A. W. et al. Metabolomic profiling of rare cell populations isolated by flow cytometry from tissues. eLife 10, e61980 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. He, S., Nakada, D. & Morrison, S. J. Mechanisms of stem cell self-renewal. Annu. Rev. Cell Dev. Biol. 25, 377–406 (2009).

    Article  CAS  PubMed  Google Scholar 

  5. Molofsky, A. V. et al. Bmi-1 dependence distinguishes neural stem cell self-renewal from progenitor proliferation. Nature 425, 962–967 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Park, I. K. et al. Bmi-1 is required for maintenance of adult self-renewing haematopoietic stem cells. Nature 423, 302–305 (2003).

    Article  CAS  PubMed  Google Scholar 

  7. Chuikov, S., Levi, B. P., Smith, M. L. & Morrison, S. J. Prdm16 promotes stem cell maintenance in multiple tissues, partly by regulating oxidative stress. Nat. Cell Biol. 12, 999–1006 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Nishino, J., Kim, I., Chada, K. & Morrison, S. J. Hmga2 promotes neural stem cell self-renewal in young but not old mice by reducing p16Ink4a and p19Arf expression. Cell 135, 227–239 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Copley, M. R. et al. The Lin28b–let-7–Hmga2 axis determines the higher self-renewal potential of fetal haematopoietic stem cells. Nat. Cell Biol. 15, 916–925 (2013).

    Article  CAS  PubMed  Google Scholar 

  10. Li, X., Egervari, G., Wang, Y., Berger, S. L. & Lu, Z. Regulation of chromatin and gene expression by metabolic enzymes and metabolites. Nat. Rev. Mol. Cell Biol. 19, 563–578 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Vander Heiden, M. G. & DeBerardinis, R. J. Understanding the intersections between metabolism and cancer biology. Cell 168, 657–669 (2017).

    Article  PubMed Central  Google Scholar 

  12. Sabari, B. R., Zhang, D., Allis, C. D. & Zhao, Y. Metabolic regulation of gene expression through histone acylations. Nat. Rev. Mol. Cell Biol. 18, 90–101 (2017).

    Article  CAS  PubMed  Google Scholar 

  13. Zhao, D. et al. Combinatorial CRISPR–Cas9 metabolic screens reveal critical redox control points dependent on the KEAP1–NRF2 regulatory axis. Mol. Cell 69, 699–708 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Wang, Y. H. et al. Cell-state-specific metabolic dependency in hematopoiesis and leukemogenesis. Cell 158, 1309–1323 (2014). This work shows that genetic changes which promote pyruvate entry into the TCA cycle impair HSC function and that this defect can be partially rescued by treatment with an antioxidant.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Takubo, K. et al. Regulation of glycolysis by Pdk functions as a metabolic checkpoint for cell cycle quiescence in hematopoietic stem cells. Cell Stem Cell 12, 49–61 (2013). This work shows that genetic changes which promote pyruvate entry into the TCA cycle impair HSC function.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Qi, L. et al. Aspartate availability limits hematopoietic stem cell function during hematopoietic regeneration. Cell Stem Cell 28, 1982–1999 (2021).

    Article  CAS  PubMed  Google Scholar 

  17. Intlekofer, A. M. & Finley, L. W. S. Metabolic signatures of cancer cells and stem cells. Nat. Metab. 1, 177–188 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  18. Burgess, R. J., Agathocleous, M. & Morrison, S. J. Metabolic regulation of stem cell function. J. Intern. Med. 276, 12–24 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Wu, J., Ocampo, A. & Belmonte, J. C. I. Cellular metabolism and induced pluripotency. Cell 166, 1371–1385 (2016).

    Article  CAS  PubMed  Google Scholar 

  20. Lu, V., Roy, I. J. & Teitell, M. A. Nutrients in the fate of pluripotent stem cells. Cell Metab. 33, 2108–2121 (2021).

    Article  CAS  PubMed  Google Scholar 

  21. Comazzetto, S., Shen, B. & Morrison, S. J. Niches that regulate stem cells and hematopoiesis in adult bone marrow. Dev. Cell 56, 1848–1860 (2021).

    Article  CAS  PubMed  Google Scholar 

  22. Spencer, J. A. et al. Direct measurement of local oxygen concentration in the bone marrow of live animals. Nature 508, 269–273 (2014). This work presents measurements of oxygen concentrations showing that HSCs reside in hypoxic regions of the bone marrow despite localizing adjacent to sinusoidal blood vessels.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Parmar, K., Mauch, P., Vergilio, J. A., Sackstein, R. & Down, J. D. Distribution of hematopoietic stem cells in the bone marrow according to regional hypoxia. Proc. Natl Acad. Sci. USA 104, 5431–5436 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Simsek, T. et al. The distinct metabolic profile of hematopoietic stem cells reflects their location in a hypoxic niche. Cell Stem Cell 7, 380–390 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Nombela-Arrieta, C. et al. Quantitative imaging of haematopoietic stem and progenitor cell localization and hypoxic status in the bone marrow microenvironment. Nat. Cell Biol. 15, 533–543 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Guitart, A. V. et al. Hif-2α is not essential for cell-autonomous hematopoietic stem cell maintenance. Blood 122, 1741–1745 (2013). This work shows that deletion of the hypoxia-inducible factors Hif1α and Hif2α has no effect on HSC function at steady state or after transplantation into irradiated mice.

    Article  CAS  PubMed  Google Scholar 

  27. Vukovic, M. et al. Adult hematopoietic stem cells lacking Hif-1α self-renew normally. Blood 127, 2841–2846 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Islam, M. S., Leissing, T. M., Chowdhury, R., Hopkinson, R. J. & Schofield, C. J. 2-Oxoglutarate-dependent oxygenases. Annu. Rev. Biochem. 87, 585–620 (2018).

    Article  CAS  PubMed  Google Scholar 

  29. Mantel, C. R. et al. Enhancing hematopoietic stem cell transplantation efficacy by mitigating oxygen shock. Cell 161, 1553–1565 (2015). This work shows that transient exposure to atmospheric oxygen levels impairs HSC function.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Mendez-Ferrer, S., Lucas, D., Battista, M. & Frenette, P. S. Haematopoietic stem cell release is regulated by circadian oscillations. Nature 452, 442–447 (2008).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  32. Morrison, S. J., Wright, D. E. & Weissman, I. L. Cyclophosphamide/granulocyte colony-stimulating factor induces hematopoietic stem cells to proliferate prior to mobilization. Proc. Natl Acad. Sci. USA 94, 1908–1913 (1997).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Jun, S. et al. The requirement for pyruvate dehydrogenase in leukemogenesis depends on cell lineage. Cell Metab. 33, 1777–1792 (2021). This work shows that HSCs take up less glucose than restricted progenitors do, suggesting that HSCs are not highly glycolytic, and PDH is dispensable for HSC function, showing that pyruvate entry into the TCA cycle is not required to maintain HSCs.

    Article  CAS  PubMed  Google Scholar 

  34. Liang, R. et al. Restraining lysosomal activity preserves hematopoietic stem cell quiescence and potency. Cell Stem Cell 26, 359–376 (2020). This work shows that activated HSCs have higher levels of lysosome activity, glycolysis and mitochondrial membrane potential than do quiescent HSCs.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Flores, A. et al. Lactate dehydrogenase activity drives hair follicle stem cell activation. Nat. Cell Biol. 19, 1017–1026 (2017).

    Article  CAS  PubMed  Google Scholar 

  36. Yucel, N. et al. Glucose metabolism drives histone acetylation landscape transitions that dictate muscle stem cell function. Cell Rep. 27, 3939–3955 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Ryall, J. G. et al. The NAD+-dependent SIRT1 deacetylase translates a metabolic switch into regulatory epigenetics in skeletal muscle stem cells. Cell Stem Cell 16, 171–183 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Sakadzic, S. et al. Two-photon high-resolution measurement of partial pressure of oxygen in cerebral vasculature and tissue. Nat. Methods 7, 755–759 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Erecińska, M. & Silver, I. A. Tissue oxygen tension and brain sensitivity to hypoxia. Respir. Physiol. 128, 263–276 (2001).

    Article  PubMed  Google Scholar 

  40. Zhang, K. et al. Reduced cerebral oxygen content in the DG and SVZ in situ promotes neurogenesis in the adult rat brain in vivo. PLoS ONE 10, e0140035 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  41. Shen, Q. et al. Adult SVZ stem cells lie in a vascular niche: a quantitative analysis of niche cell–cell interactions. Cell Stem Cell 3, 289–300 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Tavazoie, M. et al. A specialized vascular niche for adult neural stem cells. Cell Stem Cell 3, 279–288 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Palmer, T. D., Willhoite, A. R. & Gage, F. H. Vascular niche for adult hippocampal neurogenesis. J. Comp. Neurol. 425, 479–494 (2000).

    Article  CAS  PubMed  Google Scholar 

  44. Lange, C. et al. Relief of hypoxia by angiogenesis promotes neural stem cell differentiation by targeting glycolysis. EMBO J. 35, 924–994 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Yang, X., Yang, S., Wang, C. & Kuang, S. The hypoxia-inducible factors HIF1α and HIF2α are dispensable for embryonic muscle development but essential for postnatal muscle regeneration. J. Biol. Chem. 292, 5981–5991 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Moloney, J. N. & Cotter, T. G. ROS signalling in the biology of cancer. Semin. Cell Dev. Biol. 80, 50–64 (2018).

    Article  CAS  PubMed  Google Scholar 

  47. Agathocleous, M. et al. Metabolic differentiation in the embryonic retina. Nat. Cell Biol. 14, 859–864 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Khacho, M. et al. Mitochondrial dynamics impacts stem cell identity and fate decisions by regulating a nuclear transcriptional program. Cell Stem Cell 19, 232–247 (2016).

    Article  CAS  PubMed  Google Scholar 

  49. Homem, C. C. F. et al. Ecdysone and mediator change energy metabolism to terminate proliferation in Drosophila neural stem cells. Cell 158, 874–888 (2014).

    Article  CAS  PubMed  Google Scholar 

  50. van den Ameele, J. & Brand, A. H. Neural stem cell temporal patterning and brain tumour growth rely on oxidative phosphorylation. eLife 8, e47887 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  51. Martinez-Reyes, I. & Chandel, N. S. Mitochondrial TCA cycle metabolites control physiology and disease. Nat. Commun. 11, 102 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Norddahl, G. L. et al. Accumulating mitochondrial DNA mutations drive premature hematopoietic aging phenotypes distinct from physiological stem cell aging. Cell Stem Cell 8, 499–510 (2011).

    Article  CAS  PubMed  Google Scholar 

  53. de Almeida, M. J., Luchsinger, L. L., Corrigan, D. J., Williams, L. J. & Snoeck, H. W. Dye-independent methods reveal elevated mitochondrial mass in hematopoietic stem cells. Cell Stem Cell 21, 725–729 (2017). Although it was long thought that HSCs have low mitochondrial mass and activity, this study shows that HSCs have mitochondrial mass equal to or higher than that of other haematopoietic progenitors.

    Article  PubMed  PubMed Central  Google Scholar 

  54. 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 

  55. Hinge, A. et al. Asymmetrically segregated mitochondria provide cellular memory of hematopoietic stem cell replicative history and drive HSC attrition. Cell Stem Cell 26, 420–430 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Appleby, R. D. et al. Quantitation and origin of the mitochondrial membrane potential in human cells lacking mitochondrial DNA. Eur. J. Biochem. 262, 108–116 (1999).

    Article  CAS  PubMed  Google Scholar 

  57. Yahata, T. et al. Accumulation of oxidative DNA damage restricts the self-renewal capacity of human hematopoietic stem cells. Blood 118, 2941–2950 (2011).

    Article  CAS  PubMed  Google Scholar 

  58. Hu, L. et al. Antioxidant N-acetyl-l-cysteine increases engraftment of human hematopoietic stem cells in immune-deficient mice. Blood 124, e45–e48 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Ito, K. et al. Reactive oxygen species act through p38 MAPK to limit the lifespan of hematopoietic stem cells. Nat. Med. 12, 446–451 (2006).

    Article  CAS  PubMed  Google Scholar 

  60. Dixon, S. J. & Stockwell, B. R. The hallmarks of ferroptosis. Annu. Rev. Cancer Biol. 3, 35–54 (2019).

    Article  Google Scholar 

  61. Luchsinger, L. L., de Almeida, M. J., Corrigan, D. J., Mumau, M. & Snoeck, H. W. Mitofusin 2 maintains haematopoietic stem cells with extensive lymphoid potential. Nature 529, 528–531 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Jang, C., Chen, L. & Rabinowitz, J. D. Metabolomics and isotope tracing. Cell 173, 822–837 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Anso, E. et al. The mitochondrial respiratory chain is essential for haematopoietic stem cell function. Nat. Cell Biol. 19, 614–625 (2017). This work shows that deletion of a complex III electron transport chain component impairs fetal and adult HSC function, showing that HSCs depend upon the capacity for oxidative phosphorylation.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Bejarano-Garcia, J. A. et al. Sensitivity of hematopoietic stem cells to mitochondrial dysfunction by SdhD gene deletion. Cell Death Dis. 7, e2516 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Inoue, S. et al. Mitochondrial respiration defects modulate differentiation but not proliferation of hematopoietic stem and progenitor cells. FEBS Lett. 584, 3402–3409 (2010).

    Article  CAS  PubMed  Google Scholar 

  66. Yu, W. M. et al. Metabolic regulation by the mitochondrial phosphatase PTPMT1 is required for hematopoietic stem cell differentiation. Cell Stem Cell 12, 62–74 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Guitart, A. V. et al. Fumarate hydratase is a critical metabolic regulator of hematopoietic stem cell functions. J. Exp. Med. 214, 719–735 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  68. Hamanaka, R. B. et al. Mitochondrial reactive oxygen species promote epidermal differentiation and hair follicle development. Sci. Signal. 6, ra8 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  69. Kloepper, J. E. et al. Mitochondrial function in murine skin epithelium is crucial for hair follicle morphogenesis and epithelial–mesenchymal interactions. J. Invest. Dermatol. 135, 679–689 (2015).

    Article  CAS  PubMed  Google Scholar 

  70. Beckervordersandforth, R. et al. Role of mitochondrial metabolism in the control of early lineage progression and aging phenotypes in adult hippocampal neurogenesis. Neuron 93, 560–573 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  71. Zhang, F., Pirooznia, M. & Xu, H. Mitochondria regulate intestinal stem cell proliferation and epithelial homeostasis through FOXO. Mol. Biol. Cell 31, 1538–1549 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  72. Schell, J. C. et al. Control of intestinal stem cell function and proliferation by mitochondrial pyruvate metabolism. Nat. Cell Biol. 19, 1027–1036 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  73. Ito, K. et al. Regulation of oxidative stress by ATM is required for self-renewal of haematopoietic stem cells. Nature 431, 997–1002 (2004).

    Article  CAS  PubMed  Google Scholar 

  74. Tasdogan, A. et al. DNA damage-induced HSPC malfunction depends on ROS accumulation downstream of IFN-1 signaling and Bid mobilization. Cell Stem Cell 19, 752–767 (2016).

    Article  CAS  PubMed  Google Scholar 

  75. Chen, C. et al. TSC–mTOR maintains quiescence and function of hematopoietic stem cells by repressing mitochondrial biogenesis and reactive oxygen species. J. Exp. Med. 205, 2397–2408 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  76. Tothova, Z. et al. FoxOs are critical mediators of hematopoietic stem cell resistance to physiologic oxidative stress. Cell 128, 325–339 (2007).

    Article  CAS  PubMed  Google Scholar 

  77. Miyamoto, K. et al. Foxo3a is essential for maintenance of the hematopoietic stem cell pool. Cell Stem Cell 1, 101–112 (2007).

    Article  CAS  PubMed  Google Scholar 

  78. Renault, V. M. et al. FoxO3 regulates neural stem cell homeostasis. Cell Stem Cell 5, 527–539 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  79. Paik, J. H. et al. FoxOs cooperatively regulate diverse pathways governing neural stem cell homeostasis. Cell Stem Cell 5, 540–553 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  80. Yeo, H. et al. FoxO3 coordinates metabolic pathways to maintain redox balance in neural stem cells. EMBO J. 32, 2589–2602 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  81. Klotz, L. O. et al. Redox regulation of FoxO transcription factors. Redox Biol. 6, 51–72 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  82. Cuadrado, A. et al. Therapeutic targeting of the NRF2 and KEAP1 partnership in chronic diseases. Nat. Rev. Drug Discov. 18, 295–317 (2019).

    Article  CAS  PubMed  Google Scholar 

  83. Hochmuth, C. E., Biteau, B., Bohmann, D. & Jasper, H. Redox regulation by Keap1 and Nrf2 controls intestinal stem cell proliferation in Drosophila. Cell Stem Cell 8, 188–199 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  84. Holmstrom, K. M. & Finkel, T. Cellular mechanisms and physiological consequences of redox-dependent signalling. Nat. Rev. Mol. Cell Biol. 15, 411–421 (2014).

    Article  CAS  PubMed  Google Scholar 

  85. Bar-Peled, L. et al. Chemical proteomics identifies druggable vulnerabilities in a genetically defined cancer. Cell 171, 696–709 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  86. Vinogradova, E. V. et al. An activity-guided map of electrophile–cysteine interactions in primary human T cells. Cell 182, 1009–1026 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  87. 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 

  88. Covarrubias, A. J., Perrone, R., Grozio, A. & Verdin, E. NAD+ metabolism and its roles in cellular processes during ageing. Nat. Rev. Mol. Cell Biol. 22, 119–141 (2021).

    Article  CAS  PubMed  Google Scholar 

  89. 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 

  90. Stein, L. R. & Imai, S. Specific ablation of Nampt in adult neural stem cells recapitulates their functional defects during aging. EMBO J. 33, 1321–1340 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  91. Zhang, H. et al. NAD+ repletion improves mitochondrial and stem cell function and enhances life span in mice. Science 352, 1436–1443 (2016). This work shows that supplementation of mice with NAD+ precursors improves the function of stem cells in ageing tissues.

    Article  CAS  PubMed  Google Scholar 

  92. Romani, M. et al. NAD+ boosting reduces age-associated amyloidosis and restores mitochondrial homeostasis in muscle. Cell Rep. 34, 108660 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  93. Pirinen, E. et al. Niacin cures systemic NAD+ deficiency and improves muscle performance in adult-onset mitochondrial myopathy. Cell Metab. 31, 1078–1090 (2020).

    Article  CAS  PubMed  Google Scholar 

  94. 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 

  95. 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 

  96. 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 

  97. Shan, T. et al. Lkb1 is indispensable for skeletal muscle development, regeneration, and satellite cell homeostasis. Stem Cell 32, 2893–2907 (2014).

    Article  CAS  Google Scholar 

  98. Yilmaz, Ö. H. et al. Pten dependence distinguishes haematopoietic stem cells from leukaemia-initiating cells. Nature 441, 475–482 (2006).

    Article  CAS  PubMed  Google Scholar 

  99. Zhang, J. et al. PTEN maintains haematopoietic stem cells and acts in lineage choice and leukaemia prevention. Nature 441, 518–522 (2006).

    Article  CAS  PubMed  Google Scholar 

  100. Bonaguidi, M. A. et al. In vivo clonal analysis reveals self-renewing and multipotent adult neural stem cell characteristics. Cell 145, 1142–1155 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  101. Amiri, A. et al. Pten deletion in adult hippocampal neural stem/progenitor cells causes cellular abnormalities and alters neurogenesis. J. Neurosci. 32, 5880–5890 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  102. Yue, F. et al. Pten is necessary for the quiescence and maintenance of adult muscle stem cells. Nat. Commun. 8, 14328 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  103. Gan, B. et al. mTORC1-dependent and -independent regulation of stem cell renewal, differentiation, and mobilization. Proc. Natl Acad. Sci. USA 105, 19384–19389 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  104. Haller, S. et al. mTORC1 activation during repeated regeneration impairs somatic stem cell maintenance. Cell Stem Cell 21, 806–818 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  105. LaFever, L., Feoktistov, A., Hsu, H. J. & Drummond-Barbosa, D. Specific roles of target of rapamycin in the control of stem cells and their progeny in the Drosophila ovary. Development 137, 2117–2126 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  106. Wilson, A. et al. Hematopoietic stem cells reversibly switch from dormancy to self-renewal during homeostasis and repair. Cell 135, 1118–1129 (2008).

    Article  CAS  PubMed  Google Scholar 

  107. Signer, R. A., Magee, J. A., Salic, A. & Morrison, S. J. Haematopoietic stem cells require a highly regulated protein synthesis rate. Nature 509, 49–54 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  108. Blanco, S. et al. Stem cell function and stress response are controlled by protein synthesis. Nature 534, 335–340 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  109. Llorens-Bobadilla, E. et al. Single-cell transcriptomics reveals a population of dormant neural stem cells that become activated upon brain injury. Cell Stem Cell 17, 329–340 (2015).

    Article  CAS  PubMed  Google Scholar 

  110. Zismanov, V. et al. Phosphorylation of eIF2α is a translational control mechanism regulating muscle stem cell quiescence and self-renewal. Cell Stem Cell 18, 79–90 (2016).

    Article  CAS  PubMed  Google Scholar 

  111. Signer, R. A. et al. The rate of protein synthesis in hematopoietic stem cells is limited partly by 4E-BPs. Genes Dev. 30, 1698–1703 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  112. Sigurdsson, V. et al. Bile acids protect expanding hematopoietic stem cells from unfolded protein stress in fetal liver. Cell Stem Cell 18, 522–532 (2016).

    Article  CAS  PubMed  Google Scholar 

  113. Hidalgo San Jose, L. et al. Modest declines in proteome quality impair hematopoietic stem cell self-renewal. Cell Rep. 30, 69–80 (2020).

    Article  CAS  PubMed  Google Scholar 

  114. Yi, W. et al. Protein S-nitrosylation regulates proteostasis and viability of hematopoietic stem cell during regeneration. Cell Rep. 34, 108922 (2021).

    Article  CAS  PubMed  Google Scholar 

  115. Miharada, K., Sigurdsson, V. & Karlsson, S. Dppa5 improves hematopoietic stem cell activity by reducing endoplasmic reticulum stress. Cell Rep. 7, 1381–1392 (2014).

    Article  CAS  PubMed  Google Scholar 

  116. van Galen, P. et al. Integrated stress response activity marks stem cells in normal hematopoiesis and leukemia. Cell Rep. 25, 1109–1117 (2018).

    Article  PubMed  Google Scholar 

  117. Heijmans, J. et al. ER stress causes rapid loss of intestinal epithelial stemness through activation of the unfolded protein response. Cell Rep. 3, 1128–1139 (2013).

    Article  CAS  PubMed  Google Scholar 

  118. van Galen, P. et al. The unfolded protein response governs integrity of the haematopoietic stem-cell pool during stress. Nature 510, 268–272 (2014).

    Article  PubMed  Google Scholar 

  119. Buszczak, M., Signer, R. A. & Morrison, S. J. Cellular differences in protein synthesis regulate tissue homeostasis. Cell 159, 242–251 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  120. Mossmann, D., Park, S. & Hall, M. N. mTOR signalling and cellular metabolism are mutual determinants in cancer. Nat. Rev. Cancer 18, 744–757 (2018).

    Article  CAS  PubMed  Google Scholar 

  121. van Riggelen, J., Yetil, A. & Felsher, D. W. MYC as a regulator of ribosome biogenesis and protein synthesis. Nat. Rev. Cancer 10, 301–309 (2010).

    Article  PubMed  Google Scholar 

  122. Kastenhuber, E. R. & Lowe, S. W. Putting p53 in context. Cell 170, 1062–1078 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  123. Signer, R. A. & Morrison, S. J. Mechanisms that regulate stem cell aging and life span. Cell Stem Cell 12, 152–165 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  124. Pardal, R., Molofsky, A. V., He, S. & Morrison, S. J. Stem cell self-renewal and cancer cell proliferation are regulated by common networks that balance the activation of proto-oncogenes and tumor suppressors. Cold Spring Harb. Symp. Quant. Biol. 70, 177–185 (2005).

    Article  CAS  PubMed  Google Scholar 

  125. Knobloch, M. et al. Metabolic control of adult neural stem cell activity by Fasn-dependent lipogenesis. Nature 493, 226–230 (2013).

    Article  CAS  PubMed  Google Scholar 

  126. Bowers, M. et al. FASN-dependent lipid metabolism links neurogenic stem/progenitor cell activity to learning and memory deficits. Cell Stem Cell 27, 98–109 (2020).

    Article  CAS  PubMed  Google Scholar 

  127. Xie, S. Z. et al. Sphingolipid modulation activates proteostasis programs to govern human hematopoietic stem cell self-renewal. Cell Stem Cell 25, 639–653 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  128. Birsoy, K. et al. An essential role of the mitochondrial electron transport chain in cell proliferation is to enable aspartate synthesis. Cell 162, 540–551 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  129. Sullivan, L. B. et al. Supporting aspartate biosynthesis is an essential function of respiration in proliferating cells. Cell 162, 552–563 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  130. Alkan, H. F. et al. Cytosolic aspartate availability determines cell survival when glutamine is limiting. Cell Metab. 28, 706–720 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  131. Garcia-Bermudez, J. et al. Aspartate is a limiting metabolite for cancer cell proliferation under hypoxia and in tumours. Nat. Cell Biol. 20, 775–781 (2018). Together with refs 129 and 130, this study demonstrates that aspartate production within cells is limiting for their ability to survive and proliferate when electron transport chain function is undermined by hypoxia or nutritional insufficiency.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  132. Sullivan, L. B. et al. Aspartate is an endogenous metabolic limitation for tumour growth. Nat. Cell Biol. 20, 782–788 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  133. Bailis, W. et al. Distinct modes of mitochondrial metabolism uncouple T cell differentiation and function. Nature 571, 403–407 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  134. Gui, D. Y. et al. Environment dictates dependence on mitochondrial complex I for NAD+ and aspartate production and determines cancer cell sensitivity to metformin. Cell Metab. 24, 716–727 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  135. Krall, A. S. et al. Asparagine couples mitochondrial respiration to ATF4 activity and tumor growth. Cell Metab. 33, 1013–1026 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  136. Karigane, D. et al. p38α activates purine metabolism to initiate hematopoietic stem/progenitor cell cycling in response to stress. Cell Stem Cell 19, 192–204 (2016).

    Article  CAS  PubMed  Google Scholar 

  137. van Gastel, N. et al. Induction of a timed metabolic collapse to overcome cancer chemoresistance. Cell Metab. 32, 391–403 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  138. Krivtsov, A. V. et al. Transformation from committed progenitor to leukaemia stem cell initiated by MLL-AF9. Nature 442, 818–822 (2006).

    Article  CAS  PubMed  Google Scholar 

  139. Batool, T., Makky, E. A., Jalal, M. & Yusoff, M. M. A comprehensive review on l-asparaginase and its applications. Appl. Biochem. Biotechnol. 178, 900–923 (2016).

    Article  CAS  PubMed  Google Scholar 

  140. Schmiegelow, K., Nielsen, S. N., Frandsen, T. L. & Nersting, J. Mercaptopurine/methotrexate maintenance therapy of childhood acute lymphoblastic leukemia: clinical facts and fiction. J. Pediatr. Hematol. Oncol. 36, 503–517 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  141. Taya, Y. et al. Depleting dietary valine permits nonmyeloablative mouse hematopoietic stem cell transplantation. Science 354, 1152–1155 (2016). This work shows that HSCs are particularly sensitive to depletion of the branched-chain amino acid valine.

    Article  CAS  PubMed  Google Scholar 

  142. Wilkinson, A. C., Morita, M., Nakauchi, H. & Yamazaki, S. Branched-chain amino acid depletion conditions bone marrow for hematopoietic stem cell transplantation avoiding amino acid imbalance-associated toxicity. Exp. Hematol. 63, 12–16 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  143. Hattori, A. et al. Cancer progression by reprogrammed BCAA metabolism in myeloid leukaemia. Nature 545, 500–504 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  144. Gu, Z. et al. Loss of EZH2 reprograms BCAA metabolism to drive leukemic transformation. Cancer Discov. 9, 1228–1247 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  145. Raffel, S. et al. BCAT1 restricts αKG levels in AML stem cells leading to IDHmut-like DNA hypermethylation. Nature 551, 384–388 (2017).

    Article  CAS  PubMed  Google Scholar 

  146. Rodgers, J. T. et al. mTORC1 controls the adaptive transition of quiescent stem cells from G0 to G(Alert). Nature 510, 393–396 (2014). This work shows that signals induced by tissue injury cause muscle stem cells at distant sites to enter G1 phase of the cell cycle, poising them to engage in tissue regeneration, if necessary.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  147. Nakada, D. et al. Oestrogen increases haematopoietic stem-cell self-renewal in females and during pregnancy. Nature 505, 555–558 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  148. Oguro, H. et al. 27-Hydroxycholesterol induces hematopoietic stem cell mobilization and extramedullary hematopoiesis during pregnancy. J. Clin. Invest. 127, 3392–3401 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  149. Chang, N. C. Autophagy and stem cells: self-eating for self-renewal. Front. Cell Dev. Biol. 8, 138 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  150. Levine, B. & Kroemer, G. Autophagy in the pathogenesis of disease. Cell 132, 27–42 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  151. Ho, T. T. et al. Autophagy maintains the metabolism and function of young and old stem cells. Nature 543, 205–210 (2017). This work shows that autophagy promotes HSC function and that aged HSCs with high levels of autophagy have more function than that of HSCs with limited autophagy.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  152. Mortensen, M. et al. The autophagy protein Atg7 is essential for hematopoietic stem cell maintenance. J. Exp. Med. 208, 455–467 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  153. Garcia-Prat, L. et al. Autophagy maintains stemness by preventing senescence. Nature 529, 37–42 (2016).

    Article  CAS  PubMed  Google Scholar 

  154. Tang, A. H. & Rando, T. A. Induction of autophagy supports the bioenergetic demands of quiescent muscle stem cell activation. EMBO J. 33, 2782–2797 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  155. Asano, J. et al. Intrinsic autophagy is required for the maintenance of intestinal stem cells and for irradiation-induced intestinal regeneration. Cell Rep. 20, 1050–1060 (2017).

    Article  CAS  PubMed  Google Scholar 

  156. Trentesaux, C. et al. Essential role for autophagy protein ATG7 in the maintenance of intestinal stem cell integrity. Proc. Natl Acad. Sci. USA 117, 11136–11146 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  157. Kaushik, S. & Cuervo, A. M. The coming of age of chaperone-mediated autophagy. Nat. Rev. Mol. Cell Biol. 19, 365–381 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  158. Dong, S. et al. Chaperone-mediated autophagy sustains haematopoietic stem-cell function. Nature 591, 117–123 (2021). This work shows that chaperone-mediated autophagy is required for stem cell activation, increasing glycolysis and fatty acid oxidation by degrading enzymes in these pathways with deleterious post-translational modifications.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  159. Garcia-Prat, L. et al. TFEB-mediated endolysosomal activity controls human hematopoietic stem cell fate. Cell Stem Cell 28, 1–13 (2021). This work shows that lysosomal activity degrades cell surface receptors to suppress the activation of signal-transduction pathways and to prevent stem cell activation.

    Article  Google Scholar 

  160. Kobayashi, T. et al. Enhanced lysosomal degradation maintains the quiescent state of neural stem cells. Nat. Commun. 10, 5446 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  161. Leeman, D. S. et al. Lysosome activation clears aggregates and enhances quiescent neural stem cell activation during aging. Science 359, 1277–1283 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  162. Loeffler, D. et al. Asymmetric lysosome inheritance predicts activation of haematopoietic stem cells. Nature 573, 426–429 (2019).

    Article  CAS  PubMed  Google Scholar 

  163. Shook, B. A. et al. Dermal adipocyte lipolysis and myofibroblast conversion are required for efficient skin repair. Cell Stem Cell 26, 880–895 (2020). This work shows that lipolysis in adipocytes can promote tissue regeneration after injury.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  164. Zhou, B. O. et al. Bone marrow adipocytes promote the regeneration of stem cells and haematopoiesis by secreting SCF. Nat. Cell Biol. 19, 891–903 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  165. Knobloch, M. et al. A fatty acid oxidation-dependent metabolic shift regulates adult neural stem cell activity. Cell Rep. 20, 2144–2155 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  166. Mihaylova, M. M. et al. Fasting activates fatty acid oxidation to enhance intestinal stem cell function during homeostasis and aging. Cell Stem Cell 22, 769–778 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  167. Ito, K. et al. A PML–PPARδ pathway for fatty acid oxidation regulates hematopoietic stem cell maintenance. Nat. Med. 18, 1350–1358 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  168. Wellen, K. E. et al. ATP-citrate lyase links cellular metabolism to histone acetylation. Science 324, 1076–1080 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  169. Cimmino, L. et al. Restoration of TET2 function blocks aberrant self-renewal and leukemia progression. Cell 170, 1079–1095 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  170. Yue, X. & Rao, A. TET family dioxygenases and the TET activator vitamin C in immune responses and cancer. Blood 136, 1394–1401 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  171. Pan, F. et al. Tet2 loss leads to hypermutagenicity in haematopoietic stem/progenitor cells. Nat. Commun. 8, 15102 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  172. Jaiswal, S. & Ebert, B. L. Clonal hematopoiesis in human aging and disease. Science 366, eaan4673 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  173. Cabezas-Wallscheid, N. et al. Vitamin A–retinoic acid signaling regulates hematopoietic stem cell dormancy. Cell 169, 807–823 (2017).

    Article  CAS  PubMed  Google Scholar 

  174. Ghyselinck, N. B. & Duester, G. Retinoic acid signaling pathways. Development 146, dev167502 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  175. Peregrina, K. et al. Vitamin D is a determinant of mouse intestinal Lgr5 stem cell functions. Carcinogenesis 36, 25–31 (2015).

    Article  CAS  PubMed  Google Scholar 

  176. Klampfer, L. Vitamin D and colon cancer. World J. Gastrointest. Oncol. 6, 430–437 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  177. Bikle, D. & Christakos, S. New aspects of vitamin D metabolism and action — addressing the skin as source and target. Nat. Rev. Endocrinol. 16, 234–252 (2020).

    Article  CAS  PubMed  Google Scholar 

  178. Godoy-Parejo, C., Deng, C., Zhang, Y., Liu, W. & Chen, G. Roles of vitamins in stem cells. Cell Mol. Life Sci. 77, 1771–1791 (2020).

    Article  CAS  PubMed  Google Scholar 

  179. Yilmaz, O. H. et al. mTORC1 in the Paneth cell niche couples intestinal stem-cell function to calorie intake. Nature 486, 490–495 (2012). This work shows that mTORC1 activation in Paneth cells promotes an increase in the number of intestinal stem cells during caloric restriction.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  180. Igarashi, M. & Guarente, L. mTORC1 and SIRT1 cooperate to foster expansion of gut adult stem cells during calorie restriction. Cell 166, 436–450 (2016). This work shows that niche cells signal to intestinal stem cells to activate anabolic pathways during caloric restriction.

    Article  CAS  PubMed  Google Scholar 

  181. Collins, N. et al. The bone marrow protects and optimizes immunological memory during dietary restriction. Cell 178, 1088–1101 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  182. 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 

  183. Cerletti, M., Jang, Y. C., Finley, L. W., Haigis, M. C. & Wagers, A. J. Short-term calorie restriction enhances skeletal muscle stem cell function. Cell Stem Cell 10, 515–519 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  184. Forni, M. F. et al. Caloric restriction promotes structural and metabolic changes in the skin. Cell Rep. 20, 2678–2692 (2017).

    Article  CAS  PubMed  Google Scholar 

  185. Beyaz, S. et al. High-fat diet enhances stemness and tumorigenicity of intestinal progenitors. Nature 531, 53–58 (2016). This work shows that high-fat diets increase the number of stem cells in the gut epithelium, as well as tumour formation.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  186. Wang, B. et al. Phospholipid remodeling and cholesterol availability regulate intestinal stemness and tumorigenesis. Cell Stem Cell 22, 206–220 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  187. Fu, T. et al. FXR regulates intestinal cancer stem cell proliferation. Cell 176, 1098–1112 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  188. Mana, M. D. et al. High-fat diet-activated fatty acid oxidation mediates intestinal stemness and tumorigenicity. Cell Rep. 35, 109212 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  189. Morinaga, H. et al. Obesity accelerates hair thinning by stem cell-centric converging mechanisms. Nature 595, 266–271 (2021).

    Article  CAS  PubMed  Google Scholar 

  190. Cheng, C. W. et al. Ketone body signaling mediates intestinal stem cell homeostasis and adaptation to diet. Cell 178, 1115–1131 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  191. Mistry, J. J. et al. ROS-mediated PI3K activation drives mitochondrial transfer from stromal cells to hematopoietic stem cells in response to infection. Proc. Natl Acad. Sci. USA 116, 24610–24619 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  192. Golan, K. et al. Bone marrow regeneration requires mitochondrial transfer from donor Cx43-expressing hematopoietic progenitors to stroma. Blood 136, 2607–2619 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  193. Divakaruni, A. S., Paradyse, A., Ferrick, D. A., Murphy, A. N. & Jastroch, M. Analysis and interpretation of microplate-based oxygen consumption and pH data. Methods Enzymol. 547, 309–354 (2014).

    Article  CAS  PubMed  Google Scholar 

  194. Kaushik, A. K. & DeBerardinis, R. J. Applications of metabolomics to study cancer metabolism. Biochim. Biophys. Acta Rev. Cancer 1870, 2–14 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  195. Faubert, B., Tasdogan, A., Morrison, S. J., Mathews, T. P. & DeBerardinis, R. J. Stable isotope tracing to assess tumor metabolism in vivo. Nat. Protoc. 16, 5123–5145 (2021).

    Article  CAS  PubMed  Google Scholar 

  196. Lau, A. N. et al. Dissecting cell-type-specific metabolism in pancreatic ductal adenocarcinoma. eLife 9, e56782 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  197. Leveque-El Mouttie, L. et al. Autophagy is required for stem cell mobilization by G-CSF. Blood 125, 2933–2936 (2015).

    Article  CAS  PubMed  Google Scholar 

  198. Cantor, J. R. The rise of physiologic media. Trends Cell Biol. 29, 854–861 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  199. Cantor, J. R. et al. Physiologic medium rewires cellular metabolism and reveals uric acid as an endogenous inhibitor of UMP synthase. Cell 169, 258–272 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  200. Faubert, B. et al. Lactate metabolism in human lung tumors. Cell 171, 358–371 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  201. Carey, B. W., Finley, L. W., Cross, J. R., Allis, C. D. & Thompson, C. B. Intracellular α-ketoglutarate maintains the pluripotency of embryonic stem cells. Nature 518, 413–416 (2015).

    Article  CAS  PubMed  Google Scholar 

  202. Shyh-Chang, N. et al. Influence of threonine metabolism on S-adenosylmethionine and histone methylation. Science 339, 222–226 (2013).

    Article  PubMed  Google Scholar 

  203. Shiraki, N. et al. Methionine metabolism regulates maintenance and differentiation of human pluripotent stem cells. Cell Metab. 19, 780–794 (2014).

    Article  CAS  PubMed  Google Scholar 

  204. Moussaieff, A. et al. Glycolysis-mediated changes in acetyl-CoA and histone acetylation control the early differentiation of embryonic stem cells. Cell Metab. 21, 392–402 (2015).

    Article  CAS  PubMed  Google Scholar 

  205. Schieber, M. & Chandel, N. S. ROS function in redox signaling and oxidative stress. Curr. Biol. 24, 453–462 (2014).

    Article  Google Scholar 

  206. Langevin, F., Crossan, G. P., Rosado, I. V., Arends, M. J. & Patel, K. J. Fancd2 counteracts the toxic effects of naturally produced aldehydes in mice. Nature 475, 53–58 (2011).

    Article  CAS  PubMed  Google Scholar 

  207. Rosado, I. V., Langevin, F., Crossan, G. P., Takata, M. & Patel, K. J. Formaldehyde catabolism is essential in cells deficient for the Fanconi anemia DNA-repair pathway. Nat. Struct. Mol. Biol. 18, 1432–1434 (2011).

    Article  CAS  PubMed  Google Scholar 

  208. Burgos-Barragan, G. et al. Mammals divert endogenous genotoxic formaldehyde into one-carbon metabolism. Nature 548, 549–554 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  209. Garaycoechea, J. I. et al. Genotoxic consequences of endogenous aldehydes on mouse haematopoietic stem cell function. Nature 489, 571–575 (2012).

    Article  CAS  PubMed  Google Scholar 

  210. Pontel, L. B. et al. Endogenous formaldehyde is a hematopoietic stem cell genotoxin and metabolic carcinogen. Mol. Cell 60, 177–188 (2015). This work shows that detoxification of formaldehyde, a by-product of various metabolic reactions, is required to preserve genomic integrity in stem cells.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  211. Dingler, F. A. et al. Two aldehyde clearance systems are essential to prevent lethal formaldehyde accumulation in mice and humans. Mol. Cell 80, 996–1012 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

The authors thank M. Agathocleous for commenting on the manuscript and P. Mishra for discussions related to mitochondrial function. S.J.M. is a Howard Hughes Medical Institute (HHMI) Investigator, the Mary McDermott Cook Chair in Paediatric Genetics, the Kathryn and Gene Bishop Distinguished Chair in Paediatric Research, the director of the Hamon Laboratory for Stem Cells and Cancer, and a Cancer Prevention and Research Institute of Texas Scholar. This work was supported partly by the National Institutes of Health (NIH) (DK118745 to S.J.M.). C.E.M. was supported by a Postdoctoral Fellowship from the American Cancer Society (PF-13-245-01-LIB). A.W.D. was supported by a Ruth L. Kirschstein National Research Service Award Postdoctoral Fellowship from the National Heart, Lung, and Blood Institute (NHLBI) (F32HL135975).

Author information

Authors and Affiliations

Authors

Contributions

C.E.M. drafted the main text and A.W.D. drafted the figures and the glossary. S.J.M. then worked with C.E.M. to revise the text and with A.W.D. to revise the figures. All authors reviewed the manuscript before submission.

Corresponding author

Correspondence to Sean J. Morrison.

Ethics declarations

Competing interests

The authors declare no competing interests.

Peer review

Peer review information

Nature Reviews Molecular Cell Biology thanks Navdeep Chandel 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.

Supplementary information

Glossary

Hypoxia-inducible transcription factors

(HIF1α, HIF2α). Transcription factors that are degraded by the proteasome under normoxic conditions but are stabilized and translocated to the nucleus in response to low oxygen tension to promote the transcription of genes that mitigate hypoxic stress.

Ten–eleven translocation (TET) family DNA demethylases

Enzymes that mediate DNA demethylation by oxidizing 5-methylcytosine to 5-hydroxymethylcytosine.

Jumonji C histone demethylases

Enzymes that remove methyl groups from histones and other proteins.

Reactive oxygen species

(ROS). Highly reactive oxygen-containing molecules, including superoxide radicals, hydroxyl radicals and hydrogen peroxide, that are formed partly as a result of the leakage of electrons from the electron transport chain.

Mitochondrial pyruvate carrier 1

(MPC1). One subunit of the mitochondrial pyruvate transporter (MPC), which is localized to the mitochondrial inner membrane and facilitates pyruvate entry into the mitochondrial matrix.

Lactate dehydrogenase A

(LDHA). An enzyme that catalyses the reversible conversion of pyruvate and NADH to lactate and nicotinamide adenine dinucleotide (NAD+).

Sirtuin 1

(SIRT1). A nicotinamide adenine dinucleotide (NAD+)-dependent deacetylase that removes acetyl groups from histones and other proteins.

Pyruvate dehydrogenase kinase

(PDK2, PDK4). An enzyme that inactivates pyruvate dehydrogenase via phosphorylation, restricting the availability of acetyl-CoA for the tricarboxylic acid (TCA) cycle.

Dentate gyrus

A region of the hippocampus that maintains neural stem cells and neurogenesis in the adult mouse brain.

Electron transport chain

A series of protein complexes that transfer electrons within the inner mitochondrial membrane while transporting protons out of the inner mitochondrial space, creating a membrane potential. The return of these protons to the inner mitochondrial space via ATP synthase generates ATP.

Mitochondrial membrane potential

A proton gradient that is generated across the inner mitochondrial membrane as a result of electron transport chain activity. Increased mitochondrial membrane potential generally reflects increased electron transport chain activity.

Multidrug-resistance pumps

Low-specificity transporters in the plasma membrane that mediate the removal of various cytotoxic small molecules from cells.

Ferroptotic cell death

A form of cell death marked by accumulation of lipid peroxides.

Mitochondrial uncoupling agent

A small molecule or protein that dissociates mitochondrial membrane potential from ATP production by facilitating the passage of protons through the mitochondrial membrane via mechanisms independent of ATP synthase.

Extracellular flux assays

Assays performed in culture that measure the oxygen consumption rate and extracellular acidification rate of live cells in real time, providing insight into the capacity of cells to undergo oxidative phosphorylation and glycolysis.

Metabolic flux analysis

A measurement of the rate at which enzymatic reactions occur in metabolic pathways in cells, often by measuring the rates at which isotopically labelled substrates are converted into other products.

Protein mitochondrial phosphatase

(PTPMT1). A phosphatase localized to the mitochondrial inner membrane that is necessary for the biosynthesis of cardiolipin, a phospholipid that promotes the activity of many mitochondrial membrane complexes including the electron transport chain.

Fumarate hydratase 1

(FH1). An enzyme that catalyses the reversible conversion of fumarate to malate.

Pyruvate kinase M2

(PKM2). An enzyme that catalyses the last and rate-limiting step in glycolysis, the conversion of phosphoenolpyruvate to pyruvate.

Cytochrome c oxidase

(COX). An enzyme that is a component of the electron transport chain complex IV that translocates protons across the inner mitochondrial membrane, increasing mitochondrial membrane potential.

Ataxia telangiectasia mutated

(ATM). A serine/threonine kinase that is recruited to sites of DNA double-strand breaks and initiates cell cycle arrest and DNA repair.

Lysine-specific methyltransferase 2E

(KTM2E or MLL5). An atypical member of the mixed-lineage leukaemia (MLL) family that lacks methyltransferase activity but is required for DNA double-strand break repair and normal cell cycle progression.

Tuberous sclerosis complex subunit 1

(TSC1). An inhibitor of signalling by the mammalian target of rapamycin complex 1 (mTORC1).

Forkhead box O

(FOXO). A family of transcription factors that regulate the expression of genes that control many aspects of cellular and tissue homeostasis including cell cycle progression, metabolism and redox balance.

NF-E2-related factor 2

(NFE2L2 or NRF2). A transcription factor that is activated by reactive oxygen species (ROS) and that promotes the transcription of genes that encode antioxidant enzymes.

Fanconi anaemia DNA repair pathway

DNA repair proteins that are required to repair inter-strand cross links and to prevent bone marrow failure and leukaemia.

Liver kinase B1

(STK11 or LKB1). A serine/threonine kinase that activates AMP family kinases, including AMPK, a metabolic checkpoint that inhibits anabolic pathways and promotes glucose and fatty acid uptake in response to low ATP levels.

Phosphatase and tensin homologue

(PTEN). A tumour suppressor that inhibits AKT activation by dephosphorylating phosphatidylinositol triphosphate (PIP3).

4E-binding proteins

(4E-BP1, 4E-BP2). Proteins that inhibit translation by binding and inhibiting the eukaryotic translation initiation factor eIF4E.

Eukaryotic translation initiation factor 2A

(EIF2A). A subunit of the eukaryotic translation initiation factor 2 (eIF2) complex that promotes translation initiation by promoting the binding of methionine tRNAs to ribosomes and whose function is inhibited by phosphorylation.

Integrated stress response

A signalling network in eukaryotic cells that reduces protein translation and is activated by various cellular stresses including hypoxia, nutrient deprivation, accumulation of unfolded proteins and oncogene activation.

Proteotoxic stress

Cellular stress that is induced by the accumulation of misfolded or damaged proteins by heat shock, proteasome inhibition, translational infidelity, oxidative stress or accumulation of insoluble protein aggregates.

Sphingolipids

A diverse class of lipid signalling molecules that include ceramide, sphingosine and sphingosine-1-phosphate.

p38 MAP kinase

A kinase (encoded by the MAPK14 gene) that is activated by cellular stresses and pro-inflammatory cytokines.

Branched-chain aminotransferase 1

(BCAT1). An enzyme that catalyses the reversible transamination of branched-chain keto acids to the branched-chain amino acids valine, leucine and isoleucine.

Mammalian target of rapamycin complex 1

(mTORC1). A multiprotein complex that senses intracellular nutrient availability and phosphorylates substrates that promote anabolic pathways and cell cycle progression.

Fatty acid oxidation

Catabolism of fatty acids into acetyl-CoA to fuel the tricarboxylic acid (TCA) cycle and generate ATP.

Autophagy

A cellular recycling pathway in which cellular components are engulfed in a vesicle known as an autophagosome, which delivers those components to a lysosome for degradation into macromolecule precursors (that is, amino acids and nucleotides).

ATG7

An E1 ligase-like enzyme that is required for autophagosome formation.

ATG12

A ubiquitin-like modifier that is conjugated to ATG5 and is critical for the formation of the early autophagosome.

Stem cell factor

(SCF). A growth factor that is required for the maintenance of haematopoietic stem cells (HSCs) and is produced by bone marrow stromal cells, bone marrow endothelial cells and bone marrow adipocytes.

Clonal haematopoiesis

A preleukaemic condition in which a mutant clone of HSCs outcompetes other HSCs and contributes disproportionately to haematopoiesis.

Retinoic acid

The bioactive metabolite of vitamin A that mediates vitamin A functions.

Ketone bodies

Metabolites containing a ketone group that are produced from fatty acids in the liver and can be converted to acetyl-CoA for oxidation in the tricarboxylic acid (TCA) cycle.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Meacham, C.E., DeVilbiss, A.W. & Morrison, S.J. Metabolic regulation of somatic stem cells in vivo. Nat Rev Mol Cell Biol 23, 428–443 (2022). https://doi.org/10.1038/s41580-022-00462-1

Download citation

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41580-022-00462-1

This article is cited by

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