1932

Abstract

Normal tissues progressively acquire mutations. Some mutations are positively selected, driving clonal expansions that may colonize the majority of a tissue by old age. In several cases mutant clonal expansion is due to biasing stem cell fate toward proliferation. However, the expansionary phase is transient and is followed by reversion toward wild-type behavior so that normal tissue integrity is retained. Here we consider the implications of these findings for carcinogenesis. We propose that to be considered a cancer driver, a mutant gene should be more prevalent in tumors than the normal lineage from which it emerged. Cancer risk is not dependent on mutational burden, but rather may reflect the relative frequency of pro- and anti-oncogenic mutants within a tissue. Understanding the basis of mutant clonal advantage over wild-type cells allows interventions to halt the expansion or even deplete oncogenic mutants from normal tissue, potentially lowering cancer risk.

Loading

Article metrics loading...

/content/journals/10.1146/annurev-cancerbio-061421-012447
2023-04-11
2024-03-28
Loading full text...

Full text loading...

/deliver/fulltext/cancerbio/7/1/annurev-cancerbio-061421-012447.html?itemId=/content/journals/10.1146/annurev-cancerbio-061421-012447&mimeType=html&fmt=ahah

Literature Cited

  1. Abascal F, Harvey LMR, Mitchell E, Lawson ARJ, Lensing SV et al. 2021. Somatic mutation landscapes at single-molecule resolution. Nature 593:405–10
    [Google Scholar]
  2. Abby E, Dentro SC, Hall MWJ, Fowler JC, Ong SH et al. 2023. Notch1 mutation drives clonal expansion in normal esophageal epithelium but impairs tumor growth. Nat. Genet. https://doi.org/10.1038/s41588-022-01280-z
    [Google Scholar]
  3. Alcolea MP, Greulich P, Wabik A, Frede J, Simons BD, Jones PH. 2014. Differentiation imbalance in single oesophageal progenitor cells causes clonal immortalization and field change. Nat. Cell Biol. 16:615–22
    [Google Scholar]
  4. Alcolea MP, Jones PH. 2015. Cell competition: winning out by losing notch. Cell Cycle 14:9–17
    [Google Scholar]
  5. Alexandrov LB, Jones PH, Wedge DC, Sale JE, Campbell PJ et al. 2015. Clock-like mutational processes in human somatic cells. Nat. Genet. 47:1402–7
    [Google Scholar]
  6. Alexandrov LB, Ju YS, Haase K, Van Loo P, Martincorena I et al. 2016. Mutational signatures associated with tobacco smoking in human cancer. Science 354:618–22
    [Google Scholar]
  7. Alexandrov LB, Kim J, Haradhvala NJ, Huang MN, Tian Ng AW et al. 2020. The repertoire of mutational signatures in human cancer. Nature 578:94–101
    [Google Scholar]
  8. Arlt VM, Stiborova M, Schmeiser HH. 2002. Aristolochic acid as a probable human cancer hazard in herbal remedies: a review. Mutagenesis 17:265–77
    [Google Scholar]
  9. Barbera M, di Pietro M, Walker E, Brierley C, MacRae S et al. 2015. The human squamous oesophagus has widespread capacity for clonal expansion from cells at diverse stages of differentiation. Gut 64:11–19
    [Google Scholar]
  10. Barker N, van Es JH, Kuipers J, Kujala P, van den Born M et al. 2007. Identification of stem cells in small intestine and colon by marker gene Lgr5. Nature 449:1003–7
    [Google Scholar]
  11. Bick AG, Pirruccello JP, Griffin GK, Gupta N, Gabriel S et al. 2020a. Genetic interleukin 6 signaling deficiency attenuates cardiovascular risk in clonal hematopoiesis. Circulation 141:124–31
    [Google Scholar]
  12. Bick AG, Weinstock JS, Nandakumar SK, Fulco CP, Bao EL et al. 2020b. Inherited causes of clonal haematopoiesis in 97,691 whole genomes. Nature 586:763–68
    [Google Scholar]
  13. Blokzijl F, de Ligt J, Jager M, Sasselli V, Roerink S et al. 2016. Tissue-specific mutation accumulation in human adult stem cells during life. Nature 538:260–64
    [Google Scholar]
  14. Bolton KL, Ptashkin RN, Gao T, Braunstein L, Devlin SM et al. 2020. Cancer therapy shapes the fitness landscape of clonal hematopoiesis. Nat. Genet. 52:1219–26
    [Google Scholar]
  15. Bruens L, Ellenbroek SIJ, Suijkerbuijk SJE, Azkanaz M, Hale AJ et al. 2020. Calorie restriction increases the number of competing stem cells and decreases mutation retention in the intestine. Cell Rep. 32:107937
    [Google Scholar]
  16. Bruens L, Ellenbroek SIJ, van Rheenen J, Snippert HJ. 2017. In vivo imaging reveals existence of crypt fission and fusion in adult mouse intestine. Gastroenterology 153:674–77.e3
    [Google Scholar]
  17. Brunner SF, Roberts ND, Wylie LA, Moore L, Aitken SJ et al. 2019. Somatic mutations and clonal dynamics in healthy and cirrhotic human liver. Nature 574:538–42
    [Google Scholar]
  18. Challen GA, Sun D, Mayle A, Jeong M, Luo M et al. 2014. Dnmt3a and Dnmt3b have overlapping and distinct functions in hematopoietic stem cells. Cell Stem Cell 15:350–64
    [Google Scholar]
  19. Clayton E, Doupé DP, Klein AM, Winton DJ, Simons BD, Jones PH. 2007. A single type of progenitor cell maintains normal epidermis. Nature 446:185–89
    [Google Scholar]
  20. Colom B, Alcolea MP, Piedrafita G, Hall MWJ, Wabik A et al. 2020. Spatial competition shapes the dynamic mutational landscape of normal esophageal epithelium. Nat. Genet. 52:604–14
    [Google Scholar]
  21. Colom B, Herms A, Hall MWJ, Dentro SC, King C et al. 2021. Mutant clones in normal epithelium outcompete and eliminate emerging tumours. Nature 598:510–14
    [Google Scholar]
  22. Coombs CC, Zehir A, Devlin SM, Kishtagari A, Syed A et al. 2017. Therapy-related clonal hematopoiesis in patients with non-hematologic cancers is common and associated with adverse clinical outcomes. Cell Stem Cell 21:374–82.e4
    [Google Scholar]
  23. Coorens THH, Moore L, Robinson PS, Sanghvi R, Christopher J et al. 2021. Extensive phylogenies of human development inferred from somatic mutations. Nature 597:387–92
    [Google Scholar]
  24. Doupé DP, Alcolea MP, Roshan A, Zhang G, Klein AM et al. 2012. A single progenitor population switches behavior to maintain and repair esophageal epithelium. Science 337:1091–93
    [Google Scholar]
  25. Doupé DP, Klein AM, Simons BD, Jones PH. 2010. The ordered architecture of murine ear epidermis is maintained by progenitor cells with random fate. Dev. Cell 18:317–23
    [Google Scholar]
  26. Ellis P, Moore L, Sanders MA, Butler TM, Brunner SF et al. 2021. Reliable detection of somatic mutations in solid tissues by laser-capture microdissection and low-input DNA sequencing. Nat. Protoc. 16:841–71
    [Google Scholar]
  27. Fernandez-Antoran D, Piedrafita G, Murai K, Ong SH, Herms A et al. 2019. Outcompeting p53-mutant cells in the normal esophagus by redox manipulation. Cell Stem Cell 25:329–41
    [Google Scholar]
  28. Flanagan DJ, Pentinmikko N, Luopajärvi K, Willis NJ, Gilroy K et al. 2021. NOTUM from Apc-mutant cells biases clonal competition to initiate cancer. Nature 594:430–35
    [Google Scholar]
  29. Fowler JC, Jones PH. 2022. Somatic mutation: What shapes the mutational landscape of normal epithelia?. Cancer Discov. 12:1642–55
    [Google Scholar]
  30. Fowler JC, King C, Bryant C, Hall MWJ, Sood R et al. 2021. Selection of oncogenic mutant clones in normal human skin varies with body site. Cancer Discov. 11:340–61
    [Google Scholar]
  31. Fuster JJ, MacLauchlan S, Zuriaga MA, Polackal MN, Ostriker AC et al. 2017. Clonal hematopoiesis associated with TET2 deficiency accelerates atherosclerosis development in mice. Science 355:842–47
    [Google Scholar]
  32. Gallini S, Rahman N-T, Annusver K, Gonzalez DG, Yun S et al. 2022. Injury suppresses Ras cell competitive advantage through enhanced wild-type cell proliferation. bioRxiv 2022.01.05.475078. https://doi.org/10.1101/2022.01.05.475078
  33. Gerstung M, Jolly C, Leshchiner I, Dentro SC, Gonzalez S et al. 2020. The evolutionary history of 2,658 cancers. Nature 578:122–28
    [Google Scholar]
  34. Gerstung M, Papaemmanuil E, Campbell PJ. 2014. Subclonal variant calling with multiple samples and prior knowledge. Bioinformatics 30:1198–204
    [Google Scholar]
  35. Greaves LC, Preston SL, Tadrous PJ, Taylor RW, Barron MJ et al. 2006. Mitochondrial DNA mutations are established in human colonic stem cells, and mutated clones expand by crypt fission. PNAS 103:714–19
    [Google Scholar]
  36. Hauptmann M, Daniels RD, Cardis E, Cullings HM, Kendall G et al. 2020. Epidemiological studies of low-dose ionizing radiation and cancer: summary bias assessment and meta-analysis. J. Natl. Cancer Inst. Monogr. 2020:188–200
    [Google Scholar]
  37. Herms A, Bosch M, Ariotti N, Reddy Babu JN, Fajardo A et al. 2013. Cell-to-cell heterogeneity in lipid droplets suggests a mechanism to reduce lipotoxicity. Curr. Biol. 23:1489–96
    [Google Scholar]
  38. Herms A, Colom B, Piedrafita G, Murai K, Ong SH et al. 2021. Levelling out differences in aerobic glycolysis neutralizes the competitive advantage of oncogenic PIK3CA mutant progenitors in the esophagus. bioRxiv 2021.05.28.446104. https://doi.org/10.1101/2021.05.28.446104
  39. Heyde A, Rohde D, McAlpine CS, Zhang S, Hoyer FF et al. 2021. Increased stem cell proliferation in atherosclerosis accelerates clonal hematopoiesis. Cell 184:1348–61.e22
    [Google Scholar]
  40. Hill W, Zaragkoulias A, Salvador-Barbero B, Parfitt GJ, Alatsatianos M et al. 2021. EPHA2-dependent outcompetition of KRASG12D mutant cells by wild-type neighbors in the adult pancreas. Curr. Biol. 31:2550–60.e5
    [Google Scholar]
  41. Hoang ML, Chen C-H, Sidorenko VS, He J, Dickman KG et al. 2013. Mutational signature of aristolochic acid exposure as revealed by whole-exome sequencing. Sci. Transl. Med. 5:197ra102
    [Google Scholar]
  42. Hopkins BD, Goncalves MD, Cantley LC. 2020. Insulin–PI3K signalling: an evolutionarily insulated metabolic driver of cancer. Nat. Rev. Endocrinol. 16:276–83
    [Google Scholar]
  43. Hormaechea-Agulla D, Matatall KA, Le DT, Kain B, Long X et al. 2021. Chronic infection drives Dnmt3a-loss-of-function clonal hematopoiesis via IFNγ signaling. Cell Stem Cell 28:1428–42.e6
    [Google Scholar]
  44. Hsu JI, Dayaram T, Tovy A, De Braekeleer E, Jeong M et al. 2018. PPM1D mutations drive clonal hematopoiesis in response to cytotoxic chemotherapy. Cell Stem Cell 23:700–13.e6
    [Google Scholar]
  45. Jaiswal S, Natarajan P, Silver AJ, Gibson CJ, Bick AG et al. 2017. Clonal hematopoiesis and risk of atherosclerotic cardiovascular disease. New Engl. J. Med. 377:111–21
    [Google Scholar]
  46. Jha P, Ramasundarahettige C, Landsman V, Rostron B, Thun M et al. 2013. 21st-century hazards of smoking and benefits of cessation in the United States. New Engl. J. Med. 368:341–50
    [Google Scholar]
  47. Jones KB, Furukawa S, Marangoni P, Ma H, Pinkard H et al. 2019. Quantitative clonal analysis and single-cell transcriptomics reveal division kinetics, hierarchy, and fate of oral epithelial progenitor cells. Cell Stem Cell 24:183–92.e8
    [Google Scholar]
  48. Ju YS, Martincorena I, Gerstung M, Petljak M, Alexandrov LB et al. 2017. Somatic mutations reveal asymmetric cellular dynamics in the early human embryo. Nature 543:714–18
    [Google Scholar]
  49. Kahn JD, Miller PG, Silver AJ, Sellar RS, Bhatt S et al. 2018. PPM1D-truncating mutations confer resistance to chemotherapy and sensitivity to PPM1D inhibition in hematopoietic cells. Blood 132:1095–105
    [Google Scholar]
  50. Kakiuchi N, Yoshida K, Uchino M, Kihara T, Akaki K et al. 2020. Frequent mutations that converge on the NFKBIZ pathway in ulcerative colitis. Nature 577:260–65
    [Google Scholar]
  51. Klein AM, Brash DE, Jones PH, Simons BD. 2010. Stochastic fate of p53-mutant epidermal progenitor cells is tilted toward proliferation by UV B during preneoplasia. PNAS 107:270–75
    [Google Scholar]
  52. Köhnke T, Majeti R. 2021. Clonal hematopoiesis: from mechanisms to clinical intervention. Cancer Discov. 11:2987–97
    [Google Scholar]
  53. Kon S, Ishibashi K, Katoh H, Kitamoto S, Shirai T et al. 2017. Cell competition with normal epithelial cells promotes apical extrusion of transformed cells through metabolic changes. Nat. Cell Biol. 19:530–41
    [Google Scholar]
  54. Kozar S, Morrissey E, Nicholson AM, van der Heijden M, Zecchini HI et al. 2013. Continuous clonal labeling reveals small numbers of functional stem cells in intestinal crypts and adenomas. Cell Stem Cell 13:626–33
    [Google Scholar]
  55. Lawlor K, Marques-Torrejon MA, Dharmalingham G, El-Azhar Y, Schneider MD et al. 2020. Glioblastoma stem cells induce quiescence in surrounding neural stem cells via Notch signaling. Genes Dev. 34:1599–604
    [Google Scholar]
  56. Lawson ARJ, Abascal F, Coorens THH, Hooks Y, O'Neill L et al. 2020. Extensive heterogeneity in somatic mutation and selection in the human bladder. Science 370:75–82
    [Google Scholar]
  57. Lee-Six H, Olafsson S, Ellis P, Osborne RJ, Sanders MA et al. 2019. The landscape of somatic mutation in normal colorectal epithelial cells. Nature 574:532–37
    [Google Scholar]
  58. Li R, Di L, Li J, Fan W, Liu Y et al. 2021. A body map of somatic mutagenesis in morphologically normal human tissues. Nature 597:398–403
    [Google Scholar]
  59. Li R, Du Y, Chen Z, Xu D, Lin T et al. 2020. Macroscopic somatic clonal expansion in morphologically normal human urothelium. Science 370:82–89
    [Google Scholar]
  60. Lopez-Garcia C, Klein AM, Simons BD, Winton DJ. 2010. Intestinal stem cell replacement follows a neutral drift. Science 330:822–25
    [Google Scholar]
  61. Lynch MD, Lynch CNS, Craythorne E, Liakath-Ali K, Mallipeddi R et al. 2017. Spatial constraints govern competition of mutant clones in human epidermis. Nat. Commun. 8:1119
    [Google Scholar]
  62. Madissoon E, Wilbrey-Clark A, Miragaia RJ, Saeb-Parsy K, Mahbubani KT et al. 2019. scRNA-seq assessment of the human lung, spleen, and esophagus tissue stability after cold preservation. Genome Biol. 21:1
    [Google Scholar]
  63. Martincorena I, Fowler JC, Wabik A, Lawson ARJ, Abascal F et al. 2018. Somatic mutant clones colonize the human esophagus with age. Science 362:911–17
    [Google Scholar]
  64. Martincorena I, Raine KM, Gerstung M, Dawson KJ, Haase K et al. 2017. Universal patterns of selection in cancer and somatic tissues. Cell 171:1029–41.e21
    [Google Scholar]
  65. Martincorena I, Roshan A, Gerstung M, Ellis P, Van Loo P et al. 2015. High burden and pervasive positive selection of somatic mutations in normal human skin. Science 348:880–86
    [Google Scholar]
  66. Moody S, Senkin S, Islam SMA, Wang J, Nasrollahzadeh D et al. 2021. Mutational signatures in esophageal squamous cell carcinoma from eight countries with varying incidence. Nat. Genet. 53:1553–63
    [Google Scholar]
  67. Moore L, Leongamornlert D, Coorens THH, Sanders MA, Ellis P et al. 2020. The mutational landscape of normal human endometrial epithelium. Nature 580:640–46
    [Google Scholar]
  68. Murai K, Dentro S, Ong SH, Sood R, Fernandez-Antoran D et al. 2022. p53 mutation in normal esophagus promotes multiple stages of carcinogenesis but is constrained by clonal competition. Nat. Commun. 13:6206
    [Google Scholar]
  69. Murai K, Skrupskelyte G, Piedrafita G, Hall M, Kostiou V et al. 2018. Epidermal tissue adapts to restrain progenitors carrying clonal p53 mutations. Cell Stem Cell 23:687–99.e8
    [Google Scholar]
  70. Nanki K, Fujii M, Shimokawa M, Matano M, Nishikori S et al. 2020. Somatic inflammatory gene mutations in human ulcerative colitis epithelium. Nature 577:254–59
    [Google Scholar]
  71. Neuhöfer P, Roake CM, Kim SJ, Lu RJ, West RB et al. 2021. Acinar cell clonal expansion in pancreas homeostasis and carcinogenesis. Nature 597:715–19
    [Google Scholar]
  72. Ng AWT, Poon SL, Huang MN, Lim JQ, Boot A et al. 2017. Aristolochic acids and their derivatives are widely implicated in liver cancers in Taiwan and throughout Asia. Sci. Transl. Med. 9:aan6446
    [Google Scholar]
  73. Ng SWK, Rouhani FJ, Brunner SF, Brzozowska N, Aitken SJ et al. 2021. Convergent somatic mutations in metabolism genes in chronic liver disease. Nature 598:473–78
    [Google Scholar]
  74. Nicholson AM, Olpe C, Hoyle A, Thorsen AS, Rus T et al. 2018. Fixation and spread of somatic mutations in adult human colonic epithelium. Cell Stem Cell 22:909–18.e8
    [Google Scholar]
  75. Novak JSS, Baksh SC, Fuchs E. 2021. Dietary interventions as regulators of stem cell behavior in homeostasis and disease. Genes Dev. 35:199–211
    [Google Scholar]
  76. Olafsson S, McIntyre RE, Coorens T, Butler T, Jung H et al. 2020. Somatic evolution in non-neoplastic IBD-affected colon. Cell 182:672–84.e11
    [Google Scholar]
  77. Olpe C, Khamis D, Chukanova M, Skoufou-Papoutsaki N, Kemp R et al. 2021. A diffusion-like process accommodates new crypts during clonal expansion in human colonic epithelium. Gastroenterology 161:548–59.e23
    [Google Scholar]
  78. Ostrander EL, Kramer AC, Mallaney C, Celik H, Koh WK et al. 2020. Divergent effects of Dnmt3a and Tet2 mutations on hematopoietic progenitor cell fitness. Stem Cell Rep. 14:551–60
    [Google Scholar]
  79. Page ME, Lombard P, Ng F, Gottgens B, Jensen KB. 2013. The epidermis comprises autonomous compartments maintained by distinct stem cell populations. Cell Stem Cell 13:471–82
    [Google Scholar]
  80. Pareja F, Ptashkin RN, Brown DN, Derakhshan F, Selenica P et al. 2022. Cancer-causative mutations occurring in early embryogenesis. Cancer Discov. 12:949–57
    [Google Scholar]
  81. Park S, Mali NM, Kim R, Choi JW, Lee J et al. 2021. Clonal dynamics in early human embryogenesis inferred from somatic mutation. Nature 597:393–97
    [Google Scholar]
  82. Pfau SJ, Silberman RE, Knouse KA, Amon A. 2016. Aneuploidy impairs hematopoietic stem cell fitness and is selected against in regenerating tissues in vivo. Genes Dev. 30:1395–408
    [Google Scholar]
  83. Piedrafita G, Kostiou V, Wabik A, Colom B, Fernandez-Antoran D et al. 2020. A single-progenitor model as the unifying paradigm of epidermal and esophageal epithelial maintenance in mice. Nat. Commun. 11:1429
    [Google Scholar]
  84. Pineda CM, Gonzalez DG, Matte-Martone C, Boucher J, Lathrop E et al. 2019. Hair follicle regeneration suppresses Ras-driven oncogenic growth. J. Cell Biol. 218:3212–22
    [Google Scholar]
  85. Ritsma L, Ellenbroek SIJ, Zomer A, Snippert HJ, de Sauvage FJ et al. 2014. Intestinal crypt homeostasis revealed at single-stem-cell level by in vivo live imaging. Nature 507:362–65
    [Google Scholar]
  86. Riva L, Pandiri AR, Li YR, Droop A, Hewinson J et al. 2020. The mutational signature profile of known and suspected human carcinogens in mice. Nat. Genet. 52:1189–97
    [Google Scholar]
  87. Rodrigues-Moreira S, Moreno SG, Ghinatti G, Lewandowski D, Hoffschir F et al. 2017. Low-dose irradiation promotes persistent oxidative stress and decreases self-renewal in hematopoietic stem cells. Cell Rep. 20:3199–211
    [Google Scholar]
  88. Rozhok AI, Wahl GM, DeGregori J. 2015. A critical examination of the “bad luck” explanation of cancer risk. Cancer Prev. Res. 8:762–64
    [Google Scholar]
  89. Sano S, Oshima K, Wang Y, Katanasaka Y, Sano M, Walsh K. 2018. CRISPR-mediated gene editing to assess the roles of Tet2 and Dnmt3a in clonal hematopoiesis and cardiovascular disease. Circ. Res. 123:335–41
    [Google Scholar]
  90. Sasaki A, Nagatake T, Egami R, Gu G, Takigawa I et al. 2018. Obesity suppresses cell-competition-mediated apical elimination of RasV12-transformed cells from epithelial tissues. Cell Rep. 23:974–82
    [Google Scholar]
  91. Snippert HJ, Schepers AG, van Es JH, Simons BD, Clevers H. 2014. Biased competition between Lgr5 intestinal stem cells driven by oncogenic mutation induces clonal expansion. EMBO Rep. 15:62–69
    [Google Scholar]
  92. 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]
  93. Tomasetti C, Vogelstein B. 2015. Variation in cancer risk among tissues can be explained by the number of stem cell divisions. Science 347:78–81
    [Google Scholar]
  94. van Neerven SM, de Groot NE, Nijman LE, Scicluna BP, van Driel MS et al. 2021. Apc-mutant cells act as supercompetitors in intestinal tumour initiation. Nature 594:436–41
    [Google Scholar]
  95. Vermeulen L, Morrissey E, van der Heijden M, Nicholson AM, Sottoriva A et al. 2013. Defining stem cell dynamics in models of intestinal tumor initiation. Science 342:995–98
    [Google Scholar]
  96. Wabik A, Jones PH. 2015. Switching roles: the functional plasticity of adult tissue stem cells. EMBO J. 34:1164–79
    [Google Scholar]
  97. Wei L, Christensen SR, Fitzgerald ME, Graham J, Hutson ND et al. 2021. Ultradeep sequencing differentiates patterns of skin clonal mutations associated with sun-exposure status and skin cancer burden. Sci. Adv. 7:eabd7703
    [Google Scholar]
  98. Wijewardhane N, Dressler L, Ciccarelli FD. 2021. Normal somatic mutations in cancer transformation. Cancer Cell 39:125–29
    [Google Scholar]
  99. Wong SY, Reiter JF. 2011. Wounding mobilizes hair follicle stem cells to form tumors. PNAS 108:4093–98
    [Google Scholar]
  100. Yamaguchi M, Nakaoka H, Suda K, Yoshihara K, Ishiguro T et al. 2022. Spatiotemporal dynamics of clonal selection and diversification in normal endometrial epithelium. Nat. Commun. 13:943
    [Google Scholar]
  101. Yamaguchi M, Yoshihara K, Suda K, Nakaoka H, Yachida N et al. 2021. Three-dimensional understanding of the morphological complexity of the human uterine endometrium. iScience 24:102258
    [Google Scholar]
  102. Yizhak K, Aguet F, Kim J, Hess JM, Kübler K et al. 2019. RNA sequence analysis reveals macroscopic somatic clonal expansion across normal tissues. Science 364:aaw0726
    [Google Scholar]
  103. Yokoyama A, Kakiuchi N, Yoshizato T, Nannya Y, Suzuki H et al. 2019. Age-related remodelling of oesophageal epithelia by mutated cancer drivers. Nature 565:312–17
    [Google Scholar]
  104. Yoshida K, Gowers KHC, Lee-Six H, Chandrasekharan DP, Coorens T et al. 2020. Tobacco smoking and somatic mutations in human bronchial epithelium. Nature 578:266–72
    [Google Scholar]
  105. Yu H, Zhong X, Gao P, Shi J, Wu Z et al. 2019. The potential effect of metformin on cancer: an umbrella review. Front. Endocrinol. 10:617
    [Google Scholar]
  106. Yum MK, Han S, Fink J, Wu SS, Dabrowska C et al. 2021. Tracing oncogene-driven remodelling of the intestinal stem cell niche. Nature 594:442–47
    [Google Scholar]
  107. Zhang Q, Zhao K, Shen Q, Han Y, Gu Y et al. 2015. Tet2 is required to resolve inflammation by recruiting Hdac2 to specifically repress IL-6. Nature 525:389–93
    [Google Scholar]
  108. Zhang W, Remenyik E, Zelterman D, Brash DE, Wikonkal NM. 2001. Escaping the stem cell compartment: Sustained UVB exposure allows p53-mutant keratinocytes to colonize adjacent epidermal proliferating units without incurring additional mutations. PNAS 98:13948–53
    [Google Scholar]
  109. Zhu M, Lu T, Jia Y, Luo X, Gopal P et al. 2019. Somatic mutations increase hepatic clonal fitness and regeneration in chronic liver disease. Cell 177:608–21.e12
    [Google Scholar]
  110. Zink F, Stacey SN, Norddahl GL, Frigge ML, Magnusson OT et al. 2017. Clonal hematopoiesis, with and without candidate driver mutations, is common in the elderly. Blood 130:742–52
    [Google Scholar]
/content/journals/10.1146/annurev-cancerbio-061421-012447
Loading
/content/journals/10.1146/annurev-cancerbio-061421-012447
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