1932

Abstract

Although the stem cells of various tissues remain in the quiescent state to maintain their undifferentiated state, they also undergo cell divisions as required, and if necessary, even a single stem cell is able to provide for lifelong tissue homeostasis. Stem cell populations are precisely controlled by the balance between their symmetric and asymmetric divisions, with their division patterns determined by whether the daughter cells involved retain their self-renewal capacities. Recent studies have reported that metabolic pathways and the distribution of mitochondria are regulators of the division balance of stem cells and that metabolic defects can shift division balance toward symmetric commitment, which leads to stem cell exhaustion. It has also been observed that in asymmetric division, old mitochondria, which are central metabolic organelles, are segregated to the daughter cell fated to cell differentiation, whereas in symmetric division, young and old mitochondria are equally distributed between both daughter cells. Thus, metabolism and mitochondrial biology play important roles in stem cell fate decisions. As these decisions directly affect tissue homeostasis, understanding their regulatory mechanisms in the context of cellular metabolism is critical.

Loading

Article metrics loading...

/content/journals/10.1146/annurev-cellbio-111315-125134
2016-10-06
2024-03-29
Loading full text...

Full text loading...

/deliver/fulltext/cellbio/32/1/annurev-cellbio-111315-125134.html?itemId=/content/journals/10.1146/annurev-cellbio-111315-125134&mimeType=html&fmt=ahah

Literature Cited

  1. Ahn CS, Metallo CM. 2015. Mitochondria as biosynthetic factories for cancer proliferation. Cancer Metab 3:1 [Google Scholar]
  2. Appelbaum FR. 2007. Hematopoietic-cell transplantation at 50. N. Engl. J. Med. 357:1472–75 [Google Scholar]
  3. Arsenio J, Metz PJ, Chang JT. 2015. Asymmetric cell division in T lymphocyte fate diversification. Trends Immunol 36:670–83 [Google Scholar]
  4. Barbehenn EK, Wales RG, Lowry OH. 1978. Measurement of metabolites in single preimplantation embryos; a new means to study metabolic control in early embryos. J. Embryol. Exp. Morphol. 43:29–46 [Google Scholar]
  5. Beckmann J, Scheitza S, Wernet P, Fischer JC, Giebel B. 2007. Asymmetric cell division within the human hematopoietic stem and progenitor cell compartment: identification of asymmetrically segregating proteins. Blood 109:5494–501 [Google Scholar]
  6. Benveniste P, Cantin C, Hyam D, Iscove NN. 2003. Hematopoietic stem cells engraft in mice with absolute efficiency. Nat. Immunol. 4:708–13 [Google Scholar]
  7. Busch K, Klapproth K, Barile M, Flossdorf M, Holland-Letz T. et al. 2015. Fundamental properties of unperturbed haematopoiesis from stem cells in vivo. Nature 518:542–46 [Google Scholar]
  8. Carulli AJ, Samuelson LC, Schnell S. 2014. Unraveling intestinal stem cell behavior with models of crypt dynamics. Integr. Biol. Quant. Biosci. Nano Macro 6:243–57 [Google Scholar]
  9. Chandel NS. 2015. Evolution of mitochondria as signaling organelles. Cell Metab 22:204–6 [Google Scholar]
  10. Chang JT, Wherry EJ, Goldrath AW. 2014. Molecular regulation of effector and memory T cell differentiation. Nat. Immunol. 15:1104–15 [Google Scholar]
  11. Cheng T, Rodrigues N, Shen H, Yang Y, Dombkowski D. et al. 2000. Hematopoietic stem cell quiescence maintained by p21cip1/waf1. Science 287:1804–8 [Google Scholar]
  12. Cicalese A, Bonizzi G, Pasi CE, Faretta M, Ronzoni S. et al. 2009. The tumor suppressor p53 regulates polarity of self-renewing divisions in mammary stem cells. Cell 138:1083–95 [Google Scholar]
  13. Copelan EA. 2006. Hematopoietic stem-cell transplantation. N. Engl. J. Med. 354:1813–26 [Google Scholar]
  14. Daley GQ, Goodell MA, Snyder EY. 2003. Realistic prospects for stem cell therapeutics. Hematology 2003:398–418 [Google Scholar]
  15. Dykstra B, Kent D, Bowie M, McCaffrey L, Hamilton M. et al. 2007. Long-term propagation of distinct hematopoietic differentiation programs in vivo. Cell Stem Cell 1:218–29 [Google Scholar]
  16. Ema H, Takano H, Sudo K, Nakauchi H. 2000. In vitro self-renewal division of hematopoietic stem cells. J. Exp. Med. 192:1281–88 [Google Scholar]
  17. Essers MA, Offner S, Blanco-Bose WE, Waibler Z, Kalinke U. et al. 2009. IFNα activates dormant haematopoietic stem cells in vivo. Nature 458:904–8 [Google Scholar]
  18. Frenette PS, Pinho S, Lucas D, Scheiermann C. 2013. Mesenchymal stem cell: keystone of the hematopoietic stem cell niche and a stepping-stone for regenerative medicine. Annu. Rev. Immunol. 31:285–316 [Google Scholar]
  19. Gan B, Hu J, Jiang S, Liu Y, Sahin E. et al. 2010. Lkb1 regulates quiescence and metabolic homeostasis of haematopoietic stem cells. Nature 468:701–4 [Google Scholar]
  20. Gurumurthy S, Xie SZ, Alagesan B, Kim J, Yusuf RZ. et al. 2010. The Lkb1 metabolic sensor maintains haematopoietic stem cell survival. Nature 468:659–63 [Google Scholar]
  21. Ito K, Carracedo A, Weiss D, Arai F, Ala U. et al. 2012. A PML-PPAR-δ pathway for fatty acid oxidation regulates hematopoietic stem cell maintenance. Nat. Med. 18:1350–58 [Google Scholar]
  22. Ito K, Hirao A, Arai F, Matsuoka S, Takubo K. et al. 2004. Regulation of oxidative stress by ATM is required for self-renewal of haematopoietic stem cells. Nature 431:997–1002 [Google Scholar]
  23. Ito K, Hirao A, Arai F, Takubo K, Matsuoka S. et al. 2006. Reactive oxygen species act through p38 MAPK to limit the lifespan of hematopoietic stem cells. Nat. Med. 12:446–51 [Google Scholar]
  24. Ito K, Ito K. 2013. Newly identified roles of PML in stem cell biology. Front. Oncol. 3:50 [Google Scholar]
  25. Ito K, Suda T. 2014. Metabolic requirements for the maintenance of self-renewing stem cells. Nat. Rev. Mol. Cell Biol. 15:243–56 [Google Scholar]
  26. Ito T, Kwon HY, Zimdahl B, Congdon KL, Blum J. et al. 2010. Regulation of myeloid leukaemia by the cell-fate determinant Musashi. Nature 466:765–68 [Google Scholar]
  27. Katajisto P, Dohla J, Chaffer CL, Pentinmikko N, Marjanovic N. et al. 2015. Asymmetric apportioning of aged mitochondria between daughter cells is required for stemness. Science 348:340–43 [Google Scholar]
  28. Kato Y, Iwama A, Tadokoro Y, Shimoda K, Minoguchi M. et al. 2005. Selective activation of STAT5 unveils its role in stem cell self-renewal in normal and leukemic hematopoiesis. J. Exp. Med. 202:169–79 [Google Scholar]
  29. Kharas MG, Lengner CJ, Al-Shahrour F, Bullinger L, Ball B. et al. 2010. Musashi-2 regulates normal hematopoiesis and promotes aggressive myeloid leukemia. Nat. Med. 16:903–8 [Google Scholar]
  30. Kiel MJ, Yilmaz OH, Iwashita T, Yilmaz OH, Terhorst C, Morrison SJ. 2005. SLAM family receptors distinguish hematopoietic stem and progenitor cells and reveal endothelial niches for stem cells. Cell 121:1109–21 [Google Scholar]
  31. Lechler T, Fuchs E. 2005. Asymmetric cell divisions promote stratification and differentiation of mammalian skin. Nature 437:275–80 [Google Scholar]
  32. Lin WH, Adams WC, Nish SA, Chen YH, Yen B. et al. 2015. Asymmetric PI3K signaling driving developmental and regenerative cell fate bifurcation. Cell Rep 13:2203–18 [Google Scholar]
  33. Lopez-Garcia C, Klein AM, Simons BD, Winton DJ. 2010. Intestinal stem cell replacement follows a pattern of neutral drift. Science 330:822–25 [Google Scholar]
  34. Maryanovich M, Gross A. 2013. A ROS rheostat for cell fate regulation. Trends Cell Biol 23:129–34 [Google Scholar]
  35. McCulloch EA, Till JE. 1960. The radiation sensitivity of normal mouse bone marrow cells, determined by quantitative marrow transplantation into irradiated mice. Radiat. Res. 13:115–25 [Google Scholar]
  36. Miyamoto K, Araki KY, Naka K, Arai F, Takubo K. et al. 2007. Foxo3a is essential for maintenance of the hematopoietic stem cell pool. Cell Stem Cell 1:101–12 [Google Scholar]
  37. Mohrin M, Shin J, Liu Y, Brown K, Luo H. et al. 2015. A mitochondrial UPR-mediated metabolic checkpoint regulates hematopoietic stem cell aging. Science 347:1374–77 [Google Scholar]
  38. Mohyeldin A, Garzon-Muvdi T, Quinones-Hinojosa A. 2010. Oxygen in stem cell biology: a critical component of the stem cell niche. Cell Stem Cell 7:150–61 [Google Scholar]
  39. Morrison SJ, Kimble J. 2006. Asymmetric and symmetric stem-cell divisions in development and cancer. Nature 441:1068–74 [Google Scholar]
  40. Morrison SJ, Scadden DT. 2014. The bone marrow niche for haematopoietic stem cells. Nature 505:327–34 [Google Scholar]
  41. Morrison SJ, Shah NM, Anderson DJ. 1997. Regulatory mechanisms in stem cell biology. Cell 88:287–98 [Google Scholar]
  42. Nakada D, Saunders TL, Morrison SJ. 2010. Lkb1 regulates cell cycle and energy metabolism in haematopoietic stem cells. Nature 468:653–58 [Google Scholar]
  43. Osawa M, Hanada K, Hamada H, Nakauchi H. 1996. Long-term lymphohematopoietic reconstitution by a single CD34-low/negative hematopoietic stem cell. Science 273:242–45 [Google Scholar]
  44. Pagliarini DJ, Rutter J. 2013. Hallmarks of a new era in mitochondrial biochemistry. Genes Dev 27:2615–27 [Google Scholar]
  45. Panchision DM. 2009. The role of oxygen in regulating neural stem cells in development and disease. J. Cell. Physiol. 220:562–68 [Google Scholar]
  46. Piccoli C, Agriesti F, Scrima R, Falzetti F, Di Ianni M, Capitanio N. 2013. To breathe or not to breathe: the haematopoietic stem/progenitor cells dilemma. Br. J. Pharmacol. 169:1652–71 [Google Scholar]
  47. Pietras EM, Warr MR, Passegue E. 2011. Cell cycle regulation in hematopoietic stem cells. J. Cell Biol. 195:709–20 [Google Scholar]
  48. Quyn AJ, Appleton PL, Carey FA, Steele RJ, Barker N. et al. 2010. Spindle orientation bias in gut epithelial stem cell compartments is lost in precancerous tissue. Cell Stem Cell 6:175–81 [Google Scholar]
  49. Ramalho-Santos M, Willenbring H. 2007. On the origin of the term “stem cell”. Cell Stem Cell 1:35–38 [Google Scholar]
  50. Rossi DJ, Jamieson CH, Weissman IL. 2008. Stems cells and the pathways to aging and cancer. Cell 132:681–96 [Google Scholar]
  51. Scadden DT. 2006. The stem-cell niche as an entity of action. Nature 441:1075–79 [Google Scholar]
  52. Shinin V, Gayraud-Morel B, Gomes D, Tajbakhsh S. 2006. Asymmetric division and cosegregation of template DNA strands in adult muscle satellite cells. Nat. Cell Biol. 8:677–87 [Google Scholar]
  53. Shyh-Chang N, Daley GQ, Cantley LC. 2013a. Stem cell metabolism in tissue development and aging. Development 140:2535–47 [Google Scholar]
  54. Shyh-Chang N, Zhu H, Yvanka de Soysa T, Shinoda G, Seligson MT. et al. 2013b. Lin28 enhances tissue repair by reprogramming cellular metabolism. Cell 155:778–92 [Google Scholar]
  55. Simon MC, Keith B. 2008. The role of oxygen availability in embryonic development and stem cell function. Nat. Rev. Mol. Cell Biol. 9:285–96 [Google Scholar]
  56. Snippert HJ, van der Flier LG, Sato T, van Es JH, van den Born M. et al. 2010. Intestinal crypt homeostasis results from neutral competition between symmetrically dividing Lgr5 stem cells. Cell 143:134–44 [Google Scholar]
  57. Suda T, Suda J, Ogawa M. 1984. Disparate differentiation in mouse hemopoietic colonies derived from paired progenitors. PNAS 81:2520–24 [Google Scholar]
  58. Suda T, Takubo K, Semenza GL. 2011. Metabolic regulation of hematopoietic stem cells in the hypoxic niche. Cell Stem Cell 9:298–310 [Google Scholar]
  59. Sun J, Ramos A, Chapman B, Johnnidis JB, Le L. et al. 2014. Clonal dynamics of native haematopoiesis. Nature 514:322–27 [Google Scholar]
  60. Takubo K, Goda N, Yamada W, Iriuchishima H, Ikeda E. et al. 2010. Regulation of the HIF-1α level is essential for hematopoietic stem cells. Cell Stem Cell 7:391–402 [Google Scholar]
  61. Takubo K, Nagamatsu G, Kobayashi CI, Nakamura-Ishizu A, Kobayashi H. et al. 2013. Regulation of glycolysis by Pdk functions as a metabolic checkpoint for cell cycle quiescence in hematopoietic stem cells. Cell Stem Cell 12:49–61 [Google Scholar]
  62. Tothova Z, Kollipara R, Huntly BJ, Lee BH, Castrillon DH. et al. 2007. FoxOs are critical mediators of hematopoietic stem cell resistance to physiologic oxidative stress. Cell 128:325–39 [Google Scholar]
  63. Trumpp A, Essers M, Wilson A. 2010. Awakening dormant haematopoietic stem cells. Nat. Rev. Immunol. 10:201–9 [Google Scholar]
  64. Wagers AJ, Sherwood RI, Christensen JL, Weissman IL. 2002. Little evidence for developmental plasticity of adult hematopoietic stem cells. Science 297:2256–59 [Google Scholar]
  65. Walter D, Lier A, Geiselhart A, Thalheimer FB, Huntscha S. et al. 2015. Exit from dormancy provokes DNA-damage-induced attrition in haematopoietic stem cells. Nature 520:549–52 [Google Scholar]
  66. Watt FM, Hogan BL. 2000. Out of Eden: stem cells and their niches. Science 287:1427–30 [Google Scholar]
  67. Weissman IL, Anderson DJ, Gage F. 2001. Stem and progenitor cells: origins, phenotypes, lineage commitments, and transdifferentiations. Annu. Rev. Cell Dev. Biol. 17:387–403 [Google Scholar]
  68. Wilson A, Laurenti E, Oser G, van der Wath RC, Blanco-Bose W. et al. 2008. Hematopoietic stem cells reversibly switch from dormancy to self-renewal during homeostasis and repair. Cell 135:1118–29 [Google Scholar]
  69. Wu M, Kwon HY, Rattis F, Blum J, Zhao C. et al. 2007. Imaging hematopoietic precursor division in real time. Cell Stem Cell 1:541–54 [Google Scholar]
  70. Yamamoto R, Morita Y, Ooehara J, Hamanaka S, Onodera M. et al. 2013. Clonal analysis unveils self-renewing lineage-restricted progenitors generated directly from hematopoietic stem cells. Cell 154:1112–26 [Google Scholar]
  71. Yamazaki S, Iwama A, Takayanagi S, Morita Y, Eto K. et al. 2006. Cytokine signals modulated via lipid rafts mimic niche signals and induce hibernation in hematopoietic stem cells. EMBO J 25:3515–23 [Google Scholar]
  72. Yu WM, Liu X, Shen J, Jovanovic O, Pohl EE. et al. 2013. Metabolic regulation by the mitochondrial phosphatase PTPMT1 is required for hematopoietic stem cell differentiation. Cell Stem Cell 12:62–74 [Google Scholar]
  73. Zaugg K, Yao Y, Reilly PT, Kannan K, Kiarash R. et al. 2011. Carnitine palmitoyltransferase 1C promotes cell survival and tumor growth under conditions of metabolic stress. Genes Dev 25:1041–51 [Google Scholar]
  74. Zon LI. 2008. Intrinsic and extrinsic control of haematopoietic stem-cell self-renewal. Nature 453:306–13 [Google Scholar]
/content/journals/10.1146/annurev-cellbio-111315-125134
Loading
/content/journals/10.1146/annurev-cellbio-111315-125134
Loading

Data & Media loading...

  • Article Type: Review Article
This is a required field
Please enter a valid email address
Approval was a Success
Invalid data
An Error Occurred
Approval was partially successful, following selected items could not be processed due to error