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:

Deciphering the acetylation code of p53 in transcription regulation and tumor suppression

Abstract

Although it is well-established that p53-mediated tumor suppression mainly acts through its ability in transcriptional regulation, the molecular mechanisms of this regulation are not completely understood. Among a number of regulatory modes, acetylation of p53 attracts great interests. p53 was one of the first non-histone proteins found to be functionally regulated by acetylation and deacetylation, and subsequent work has established that reversible acetylation is a general mechanism for regulation of non-histone proteins. Unlike other types of posttranslational modifications occurred during stress responses, the role of p53 acetylation has been recently validated in vivo by using the knock-in mice with both acetylation-defective and acetylation-mimicking p53 mutants. Here, we review the role of acetylation in p53-mediated activities, with a focus on which specific acetylation sites are critical for p53-dependent transcription regulation during tumor suppression and how acetylation of p53 recruits specific “readers” to execute its promoter-specific regulation of different targets. We also discuss the role of p53 acetylation in differentially regulating its classic activities in cell cycle arrest, senescence and apoptosis as well as newly identified unconventional functions such as cell metabolism and ferroptosis.

This is a preview of subscription content, access via your institution

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

Prices may be subject to local taxes which are calculated during checkout

Fig. 1: Overview of p53 acetylation sites and enzymes involved.
Fig. 2: Functional significance of p53 acetylation underlined by mouse models.
Fig. 3: Graphical presentation of acetylation-mediated regulation of p53 tumor suppressive functions, as revealed in mouse models.
Fig. 4: Distinct functional consequences of p53 acetylation in DBD and CTD.
Fig. 5: Molecular readers for p53 acetylation.

Similar content being viewed by others

References

  1. Boutelle AM, Attardi LD. p53 and tumor suppression: it takes a network. Trends Cell Biol. 2021;31:298–310.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Mello SS, Attardi LD. Deciphering p53 signaling in tumor suppression. Curr Opin Cell Biol. 2018;51:65–72.

    Article  CAS  PubMed  Google Scholar 

  3. Kaiser AM, Attardi LD. Deconstructing networks of p53-mediated tumor suppression in vivo. Cell Death Differ. 2018;25:93–103.

    Article  CAS  PubMed  Google Scholar 

  4. Liu Y, Tavana O, Gu W. p53 modifications: exquisite decorations of the powerful guardian. J Mol Cell Biol. 2019;11:564–77.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Gu W, Roeder RG. Activation of p53 sequence-specific DNA binding by acetylation of the p53 C-terminal domain. Cell. 1997;90:595–606.

    Article  CAS  PubMed  Google Scholar 

  6. Tang Y, Zhao W, Chen Y, Zhao Y, Gu W. Acetylation is indispensable for p53 activation. Cell. 2008;133:612–26.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Barnoud T, Indeglia A, Murphy ME. Shifting the paradigms for tumor suppression: lessons from the p53 field. Oncogene. 2021;40:4281–90.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Reed SM, Quelle DE. p53 acetylation: regulation and consequences. Cancers. 2014;7:30–69.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  9. Wen J, Wang D. Deciphering the PTM codes of the tumor suppressor p53. J Mol Cell Biol. 2022;13:774–85.

  10. Allfrey VG, Faulkner R, Mirsky AE. Acetylation and methylation of histones and their possible role in the regulation of Rna synthesis. Proc Natl Acad Sci USA. 1964;51:786–94.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Nitsch S, Zorro Shahidian L, Schneider R. Histone acylations and chromatin dynamics: concepts, challenges, and links to metabolism. EMBO Rep. 2021;22:e52774.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Sterner R, Vidali G, Allfrey VG. Studies of acetylation and deacetylation in high mobility group proteins. Identification of the sites of acetylation in HMG-1. J Biol Chem. 1979;254:11577–83.

    Article  CAS  PubMed  Google Scholar 

  13. L’Hernault SW, Rosenbaum JL. Chlamydomonas alpha-tubulin is posttranslationally modified by acetylation on the epsilon-amino group of a lysine. Biochemistry. 1985;24:473–8.

    Article  PubMed  Google Scholar 

  14. Gu W, Shi XL, Roeder RG. Synergistic activation of transcription by CBP and p53. Nature. 1997;387:819–23.

    Article  CAS  PubMed  Google Scholar 

  15. Tang Y, Luo J, Zhang W, Gu W. Tip60-dependent acetylation of p53 modulates the decision between cell-cycle arrest and apoptosis. Mol Cell. 2006;24:827–39.

    Article  CAS  PubMed  Google Scholar 

  16. Li T, Kon N, Jiang L, Tan M, Ludwig T, Zhao Y, et al. Tumor suppression in the absence of p53-mediated cell-cycle arrest, apoptosis, and senescence. Cell. 2012;149:1269–83.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Jiang L, Kon N, Li T, Wang SJ, Su T, Hibshoosh H, et al. Ferroptosis as a p53-mediated activity during tumour suppression. Nature. 2015;520:57–62.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Wang SJ, Li D, Ou Y, Jiang L, Chen Y, Zhao Y, et al. Acetylation is crucial for p53-mediated ferroptosis and tumor suppression. Cell Rep. 2016;17:366–73.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Kon N, Ou Y, Wang SJ, Li H, Rustgi AK, Gu W. Corrigendum: mTOR inhibition acts as an unexpected checkpoint in p53-mediated tumor suppression. Genes Dev. 2021;35:300.

    Article  PubMed  PubMed Central  Google Scholar 

  20. Wang D, Kon N, Lasso G, Jiang L, Leng W, Zhu WG, et al. Acetylation-regulated interaction between p53 and SET reveals a widespread regulatory mode. Nature. 2016;538:118–22.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Wang D, Kon N, Tavana O, Gu W. The “readers” of unacetylated p53 represent a new class of acidic domain proteins. Nucleus. 2017;8:360–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Yang X, Wang X, Li Z, Duan S, Li H, Jin J, et al. An unexpected role for Dicer as a reader of the unacetylated DNA binding domain of p53 in transcriptional regulation. Sci Adv. 2021;7:eabi6684.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Choudhary C, Kumar C, Gnad F, Nielsen ML, Rehman M, Walther TC, et al. Lysine acetylation targets protein complexes and co-regulates major cellular functions. Science. 2009;325:834–40.

    Article  CAS  PubMed  Google Scholar 

  24. Narita T, Weinert BT, Choudhary C. Functions and mechanisms of non-histone protein acetylation. Nat Rev Mol Cell Biol. 2019;20:156–74.

    Article  CAS  PubMed  Google Scholar 

  25. Verdin E, Ott M. 50 years of protein acetylation: from gene regulation to epigenetics, metabolism and beyond. Nat Rev Mol Cell Biol. 2015;16:258–64.

    Article  CAS  PubMed  Google Scholar 

  26. Joerger AC, Fersht AR. The p53 pathway: origins, inactivation in cancer, and emerging therapeutic approaches. Annu Rev Biochem. 2016;85:375–404.

    Article  CAS  PubMed  Google Scholar 

  27. Avantaggiati ML, Ogryzko V, Gardner K, Giordano A, Levine AS, Kelly K. Recruitment of p300/CBP in p53-dependent signal pathways. Cell. 1997;89:1175–84.

    Article  CAS  PubMed  Google Scholar 

  28. Lill NL, Grossman SR, Ginsberg D, DeCaprio J, Livingston DM. Binding and modulation of p53 by p300/CBP coactivators. Nature. 1997;387:823–7.

    Article  CAS  PubMed  Google Scholar 

  29. Scolnick DM, Chehab NH, Stavridi ES, Lien MC, Caruso L, Moran E, et al. CREB-binding protein and p300/CBP-associated factor are transcriptional coactivators of the p53 tumor suppressor protein. Cancer Res. 1997;57:3693–6.

    CAS  PubMed  Google Scholar 

  30. Grossman SR, Perez M, Kung AL, Joseph M, Mansur C, Xiao ZX, et al. p300/MDM2 complexes participate in MDM2-mediated p53 degradation. Mol Cell. 1998;2:405–15.

    Article  CAS  PubMed  Google Scholar 

  31. Wadgaonkar R, Phelps KM, Haque Z, Williams AJ, Silverman ES, Collins T. CREB-binding protein is a nuclear integrator of nuclear factor-kappaB and p53 signaling. J Biol Chem. 1999;274:1879–82.

    Article  CAS  PubMed  Google Scholar 

  32. Buschmann T, Adler V, Matusevich E, Fuchs SY, Ronai Z. p53 phosphorylation and association with murine double minute 2, c-Jun NH2-terminal kinase, p14ARF, and p300/CBP during the cell cycle and after exposure to ultraviolet irradiation. Cancer Res. 2000;60:896–900.

    CAS  PubMed  Google Scholar 

  33. Yang XJ, Ogryzko VV, Nishikawa J, Howard BH, Nakatani Y. A p300/CBP-associated factor that competes with the adenoviral oncoprotein E1A. Nature. 1996;382:319–24.

    Article  CAS  PubMed  Google Scholar 

  34. Liu L, Scolnick DM, Trievel RC, Zhang HB, Marmorstein R, Halazonetis TD, et al. p53 sites acetylated in vitro by PCAF and p300 are acetylated in vivo in response to DNA damage. Mol Cell Biol. 1999;19:1202–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Sakaguchi K, Herrera JE, Saito S, Miki T, Bustin M, Vassilev A, et al. DNA damage activates p53 through a phosphorylation-acetylation cascade. Genes Dev. 1998;12:2831–41.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Sykes SM, Mellert HS, Holbert MA, Li K, Marmorstein R, Lane WS, et al. Acetylation of the p53 DNA-binding domain regulates apoptosis induction. Mol Cell. 2006;24:841–51.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Li X, Wu L, Corsa CA, Kunkel S, Dou Y. Two mammalian MOF complexes regulate transcription activation by distinct mechanisms. Mol Cell. 2009;36:290–301.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Rokudai S, Laptenko O, Arnal SM, Taya Y, Kitabayashi I, Prives C. MOZ increases p53 acetylation and premature senescence through its complex formation with PML. Proc Natl Acad Sci USA. 2013;110:3895–900.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Wang YH, Tsay YG, Tan BC, Lo WY, Lee SC. Identification and characterization of a novel p300-mediated p53 acetylation site, lysine 305. J Biol Chem. 2003;278:25568–76.

    Article  CAS  PubMed  Google Scholar 

  40. Joubel A, Chalkley RJ, Medzihradszky KF, Hondermarck H, Burlingame AL. Identification of new p53 acetylation sites in COS-1 cells. Mol Cell Proteom. 2009;8:1167–73.

    Article  CAS  Google Scholar 

  41. Luo J, Su F, Chen D, Shiloh A, Gu W. Deacetylation of p53 modulates its effect on cell growth and apoptosis. Nature. 2000;408:377–81.

    Article  CAS  PubMed  Google Scholar 

  42. Luo J, Nikolaev AY, Imai S, Chen D, Su F, Shiloh A, et al. Negative control of p53 by Sir2alpha promotes cell survival under stress. Cell. 2001;107:137–48.

    Article  CAS  PubMed  Google Scholar 

  43. Vaziri H, Dessain SK, Ng Eaton E, Imai SI, Frye RA, Pandita TK, et al. hSIR2(SIRT1) functions as an NAD-dependent p53 deacetylase. Cell. 2001;107:149–59.

    Article  CAS  PubMed  Google Scholar 

  44. Feng L, Lin T, Uranishi H, Gu W, Xu Y. Functional analysis of the roles of posttranslational modifications at the p53 C terminus in regulating p53 stability and activity. Mol Cell Biol. 2005;25:5389–95.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Krummel KA, Lee CJ, Toledo F, Wahl GM. The C-terminal lysines fine-tune P53 stress responses in a mouse model but are not required for stability control or transactivation. Proc Natl Acad Sci USA. 2005;102:10188–93.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Simeonova I, Jaber S, Draskovic I, Bardot B, Fang M, Bouarich-Bourimi R, et al. Mutant mice lacking the p53 C-terminal domain model telomere syndromes. Cell Rep. 2013;3:2046–58.

    Article  CAS  PubMed  Google Scholar 

  47. Hamard PJ, Barthelery N, Hogstad B, Mungamuri SK, Tonnessen CA, Carvajal LA, et al. The C terminus of p53 regulates gene expression by multiple mechanisms in a target- and tissue-specific manner in vivo. Genes Dev. 2013;27:1868–85.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Hupp TR, Meek DW, Midgley CA, Lane DP. Regulation of the specific DNA binding function of p53. Cell. 1992;71:875–86.

    Article  CAS  PubMed  Google Scholar 

  49. Hupp TR, Sparks A, Lane DP. Small peptides activate the latent sequence-specific DNA binding function of p53. Cell. 1995;83:237–45.

    Article  CAS  PubMed  Google Scholar 

  50. Jayaraman J, Prives C. Activation of p53 sequence-specific DNA binding by short single strands of DNA requires the p53 C-terminus. Cell. 1995;81:1021–9.

    Article  CAS  PubMed  Google Scholar 

  51. Kon N, Churchill M, Li H, Mukherjee S, Manfredi JJ, Gu W. Robust p53 stabilization is dispensable for its activation and tumor suppressor function. Cancer Res. 2021;81:935–44.

    Article  CAS  PubMed  Google Scholar 

  52. Kon N, Gu W. p53 activation vs. stabilization: an acetylation tale from the C-terminal tail. Oncoscience. 2021;8:58–60.

    Article  PubMed  PubMed Central  Google Scholar 

  53. Knights CD, Catania J, Di Giovanni S, Muratoglu S, Perez R, Swartzbeck A, et al. Distinct p53 acetylation cassettes differentially influence gene-expression patterns and cell fate. J Cell Biol. 2006;173:533–44.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Chao C, Wu Z, Mazur SJ, Borges H, Rossi M, Lin T, et al. Acetylation of mouse p53 at lysine 317 negatively regulates p53 apoptotic activities after DNA damage. Mol Cell Biol. 2006;26:6859–69.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Montes de Oca Luna R, Wagner DS, Lozano G. Rescue of early embryonic lethality in mdm2-deficient mice by deletion of p53. Nature. 1995;378:203–6.

    Article  CAS  PubMed  Google Scholar 

  56. Kon N, Wang D, Li T, Jiang L, Qiang L, Gu W. Inhibition of Mdmx (Mdm4) in vivo induces anti-obesity effects. Oncotarget. 2018;9:7282–97.

    Article  PubMed  PubMed Central  Google Scholar 

  57. Brady CA, Jiang D, Mello SS, Johnson TM, Jarvis LA, Kozak MM, et al. Distinct p53 transcriptional programs dictate acute DNA-damage responses and tumor suppression. Cell. 2011;145:571–83.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Valente LJ, Gray DH, Michalak EM, Pinon-Hofbauer J, Egle A, Scott CL, et al. p53 efficiently suppresses tumor development in the complete absence of its cell-cycle inhibitory and proapoptotic effectors p21, Puma, and Noxa. Cell Rep. 2013;3:1339–45.

    Article  CAS  PubMed  Google Scholar 

  59. Li T, Liu X, Jiang L, Manfredi J, Zha S, Gu W. Loss of p53-mediated cell-cycle arrest, senescence and apoptosis promotes genomic instability and premature aging. Oncotarget. 2016;7:11838–49.

    Article  PubMed  PubMed Central  Google Scholar 

  60. Liu Y, Gu W. The complexity of p53-mediated metabolic regulation in tumor suppression. Semin Cancer Biol. 2021.

  61. Bensaad K, Tsuruta A, Selak MA, Vidal MN, Nakano K, Bartrons R, et al. TIGAR, a p53-inducible regulator of glycolysis and apoptosis. Cell. 2006;126:107–20.

    Article  CAS  PubMed  Google Scholar 

  62. Cheung EC, Ludwig RL, Vousden KH. Mitochondrial localization of TIGAR under hypoxia stimulates HK2 and lowers ROS and cell death. Proc Natl Acad Sci USA. 2012;109:20491–6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Cheung EC, Athineos D, Lee P, Ridgway RA, Lambie W, Nixon C, et al. TIGAR is required for efficient intestinal regeneration and tumorigenesis. Dev Cell. 2013;25:463–77.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Cheung EC, DeNicola GM, Nixon C, Blyth K, Labuschagne CF, Tuveson DA, et al. Dynamic ROS control by TIGAR regulates the initiation and progression of pancreatic cancer. Cancer Cell. 2020;37:168–82 e4.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Assi M. The differential role of reactive oxygen species in early and late stages of cancer. Am J Physiol Regul Integr Comp Physiol. 2017;313:R646–53.

    Article  PubMed  CAS  Google Scholar 

  66. Stockwell BR, Jiang X, Gu W. Emerging mechanisms and disease relevance of ferroptosis. Trends Cell Biol. 2020;30:478–90.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Yang WS, Stockwell BR. Ferroptosis: death by lipid peroxidation. Trends Cell Biol. 2016;26:165–76.

    Article  CAS  PubMed  Google Scholar 

  68. Jiang L, Hickman JH, Wang SJ, Gu W. Dynamic roles of p53-mediated metabolic activities in ROS-induced stress responses. Cell Cycle. 2015;14:2881–5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  69. Jennis M, Kung CP, Basu S, Budina-Kolomets A, Leu JI, Khaku S, et al. An African-specific polymorphism in the TP53 gene impairs p53 tumor suppressor function in a mouse model. Genes Dev. 2016;30:918–30.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  70. Leu JI, Murphy ME, George DL. Mechanistic basis for impaired ferroptosis in cells expressing the African-centric S47 variant of p53. Proc Natl Acad Sci USA. 2019;116:8390–6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  71. Badgley MA, Kremer DM, Maurer HC, DelGiorno KE, Lee HJ, Purohit V, et al. Cysteine depletion induces pancreatic tumor ferroptosis in mice. Science. 2020;368:85–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  72. Chu B, Kon N, Chen D, Li T, Liu T, Jiang L, et al. ALOX12 is required for p53-mediated tumour suppression through a distinct ferroptosis pathway. Nat Cell Biol. 2019;21:579–91.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  73. Doll S, Proneth B, Tyurina YY, Panzilius E, Kobayashi S, Ingold I, et al. ACSL4 dictates ferroptosis sensitivity by shaping cellular lipid composition. Nat Chem Biol. 2017;13:91–8.

    Article  CAS  PubMed  Google Scholar 

  74. Kagan VE, Mao G, Qu F, Angeli JP, Doll S, Croix CS, et al. Oxidized arachidonic and adrenic PEs navigate cells to ferroptosis. Nat Chem Biol. 2017;13:81–90.

    Article  CAS  PubMed  Google Scholar 

  75. Gao M, Monian P, Quadri N, Ramasamy R, Jiang X. Glutaminolysis and transferrin regulate ferroptosis. Mol Cell. 2015;59:298–308.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  76. Ou Y, Wang SJ, Li D, Chu B, Gu W. Activation of SAT1 engages polyamine metabolism with p53-mediated ferroptotic responses. Proc Natl Acad Sci USA. 2016;113:E6806–12.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  77. Zhang Y, Qian Y, Zhang J, Yan W, Jung YS, Chen M, et al. Ferredoxin reductase is critical for p53-dependent tumor suppression via iron regulatory protein 2. Genes Dev. 2017;31:1243–56.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  78. Liu Y, Gu W. p53 in ferroptosis regulation: the new weapon for the old guardian. Cell Death Differ. 2022.

  79. Li M, Luo J, Brooks CL, Gu W. Acetylation of p53 inhibits its ubiquitination by Mdm2. J Biol Chem. 2002;277:50607–11.

    Article  CAS  PubMed  Google Scholar 

  80. Kruse JP, Gu W. Modes of p53 regulation. Cell. 2009;137:609–22.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  81. Rodriguez MS, Desterro JM, Lain S, Lane DP, Hay RT. Multiple C-terminal lysine residues target p53 for ubiquitin-proteasome-mediated degradation. Mol Cell Biol. 2000;20:8458–67.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  82. Ito A, Lai CH, Zhao X, Saito S, Hamilton MH, Appella E, et al. p300/CBP-mediated p53 acetylation is commonly induced by p53-activating agents and inhibited by MDM2. EMBO J. 2001;20:1331–40.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  83. Kobet E, Zeng X, Zhu Y, Keller D, Lu H. MDM2 inhibits p300-mediated p53 acetylation and activation by forming a ternary complex with the two proteins. Proc Natl Acad Sci USA. 2000;97:12547–52.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  84. Ito A, Kawaguchi Y, Lai CH, Kovacs JJ, Higashimoto Y, Appella E, et al. MDM2-HDAC1-mediated deacetylation of p53 is required for its degradation. EMBO J. 2002;21:6236–45.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  85. Fujisawa T, Filippakopoulos P. Functions of bromodomain-containing proteins and their roles in homeostasis and cancer. Nat Rev Mol Cell Biol. 2017;18:246–62.

    Article  CAS  PubMed  Google Scholar 

  86. Barlev NA, Liu L, Chehab NH, Mansfield K, Harris KG, Halazonetis TD, et al. Acetylation of p53 activates transcription through recruitment of coactivators/histone acetyltransferases. Mol Cell. 2001;8:1243–54.

    Article  CAS  PubMed  Google Scholar 

  87. Li AG, Piluso LG, Cai X, Gadd BJ, Ladurner AG, Liu X. An acetylation switch in p53 mediates holo-TFIID recruitment. Mol Cell. 2007;28:408–21.

    Article  PubMed  CAS  Google Scholar 

  88. Cai W, Su L, Liao L, Liu ZZ, Langbein L, Dulaimi E, et al. PBRM1 acts as a p53 lysine-acetylation reader to suppress renal tumor growth. Nat Commun. 2019;10:5800.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  89. Juillard F, de Miranda MP, Li S, Franco A, Seixas AF, Liu B, et al. KSHV LANA acetylation-selective acidic domain reader sequence mediates virus persistence. Proc Natl Acad Sci USA. 2020;117:22443–51.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  90. Meek DW, Anderson CW. Posttranslational modification of p53: cooperative integrators of function. Cold Spring Harb Perspect Biol. 2009;1:a000950.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  91. Lambert PF, Kashanchi F, Radonovich MF, Shiekhattar R, Brady JN. Phosphorylation of p53 serine 15 increases interaction with CBP. J Biol Chem. 1998;273:33048–53.

    Article  CAS  PubMed  Google Scholar 

  92. Kar S, Sakaguchi K, Shimohigashi Y, Samaddar S, Banerjee R, Basu G, et al. Effect of phosphorylation on the structure and fold of transactivation domain of p53. J Biol Chem. 2002;277:15579–85.

    Article  CAS  PubMed  Google Scholar 

  93. Teufel DP, Bycroft M, Fersht AR. Regulation by phosphorylation of the relative affinities of the N-terminal transactivation domains of p53 for p300 domains and Mdm2. Oncogene. 2009;28:2112–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  94. Jenkins LM, Yamaguchi H, Hayashi R, Cherry S, Tropea JE, Miller M, et al. Two distinct motifs within the p53 transactivation domain bind to the Taz2 domain of p300 and are differentially affected by phosphorylation. Biochemistry. 2009;48:1244–55.

    Article  PubMed  CAS  Google Scholar 

  95. Lee CW, Ferreon JC, Ferreon AC, Arai M, Wright PE. Graded enhancement of p53 binding to CREB-binding protein (CBP) by multisite phosphorylation. Proc Natl Acad Sci USA. 2010;107:19290–5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  96. Saito S, Goodarzi AA, Higashimoto Y, Noda Y, Lees-Miller SP, Appella E, et al. ATM mediates phosphorylation at multiple p53 sites, including Ser(46), in response to ionizing radiation. J Biol Chem. 2002;277:12491–4.

    Article  CAS  PubMed  Google Scholar 

  97. Hofmann TG, Moller A, Sirma H, Zentgraf H, Taya Y, Droge W, et al. Regulation of p53 activity by its interaction with homeodomain-interacting protein kinase-2. Nat Cell Biol. 2002;4:1–10.

    Article  CAS  PubMed  Google Scholar 

  98. Puca R, Nardinocchi L, Sacchi A, Rechavi G, Givol D, D’Orazi G. HIPK2 modulates p53 activity towards pro-apoptotic transcription. Mol Cancer. 2009;8:85.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  99. Rokudai S, Aikawa Y, Tagata Y, Tsuchida N, Taya Y, Kitabayashi I. Monocytic leukemia zinc finger (MOZ) interacts with p53 to induce p21 expression and cell-cycle arrest. J Biol Chem. 2009;284:237–44.

    Article  CAS  PubMed  Google Scholar 

  100. Shieh SY, Ikeda M, Taya Y, Prives C. DNA damage-induced phosphorylation of p53 alleviates inhibition by MDM2. Cell. 1997;91:325–34.

    Article  CAS  PubMed  Google Scholar 

  101. Chehab NH, Malikzay A, Appel M, Halazonetis TD. Chk2/hCds1 functions as a DNA damage checkpoint in G(1) by stabilizing p53. Genes Dev. 2000;14:278–88.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  102. Ferreon JC, Lee CW, Arai M, Martinez-Yamout MA, Dyson HJ, Wright PE. Cooperative regulation of p53 by modulation of ternary complex formation with CBP/p300 and HDM2. Proc Natl Acad Sci USA. 2009;106:6591–6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  103. Ou YH, Chung PH, Sun TP, Shieh SY. p53 C-terminal phosphorylation by CHK1 and CHK2 participates in the regulation of DNA-damage-induced C-terminal acetylation. Mol Biol Cell. 2005;16:1684–95.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  104. Chuikov S, Kurash JK, Wilson JR, Xiao B, Justin N, Ivanov GS, et al. Regulation of p53 activity through lysine methylation. Nature. 2004;432:353–60.

    Article  CAS  PubMed  Google Scholar 

  105. Huang J, Perez-Burgos L, Placek BJ, Sengupta R, Richter M, Dorsey JA, et al. Repression of p53 activity by Smyd2-mediated methylation. Nature. 2006;444:629–32.

    Article  CAS  PubMed  Google Scholar 

  106. Shi X, Kachirskaia I, Yamaguchi H, West LE, Wen H, Wang EW, et al. Modulation of p53 function by SET8-mediated methylation at lysine 382. Mol Cell. 2007;27:636–46.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  107. Ivanov GS, Ivanova T, Kurash J, Ivanov A, Chuikov S, Gizatullin F, et al. Methylation-acetylation interplay activates p53 in response to DNA damage. Mol Cell Biol. 2007;27:6756–69.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  108. Kurash JK, Lei H, Shen Q, Marston WL, Granda BW, Fan H, et al. Methylation of p53 by Set7/9 mediates p53 acetylation and activity in vivo. Mol Cell. 2008;29:392–400.

    Article  CAS  PubMed  Google Scholar 

  109. Lehnertz B, Rogalski JC, Schulze FM, Yi L, Lin S, Kast J, et al. p53-dependent transcription and tumor suppression are not affected in Set7/9-deficient mice. Mol Cell. 2011;43:673–80.

    Article  CAS  PubMed  Google Scholar 

  110. Campaner S, Spreafico F, Burgold T, Doni M, Rosato U, Amati B, et al. The methyltransferase Set7/9 (Setd7) is dispensable for the p53-mediated DNA damage response in vivo. Mol Cell. 2011;43:681–8.

    Article  CAS  PubMed  Google Scholar 

  111. Rodriguez MS, Desterro JM, Lain S, Midgley CA, Lane DP, Hay RT. SUMO-1 modification activates the transcriptional response of p53. EMBO J. 1999;18:6455–61.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  112. Gostissa M, Hengstermann A, Fogal V, Sandy P, Schwarz SE, Scheffner M, et al. Activation of p53 by conjugation to the ubiquitin-like protein SUMO-1. EMBO J. 1999;18:6462–71.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  113. Fogal V, Gostissa M, Sandy P, Zacchi P, Sternsdorf T, Jensen K, et al. Regulation of p53 activity in nuclear bodies by a specific PML isoform. EMBO J. 2000;19:6185–95.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  114. Kwek SS, Derry J, Tyner AL, Shen Z, Gudkov AV. Functional analysis and intracellular localization of p53 modified by SUMO-1. Oncogene. 2001;20:2587–99.

    Article  CAS  PubMed  Google Scholar 

  115. Carter S, Bischof O, Dejean A, Vousden KH. C-terminal modifications regulate MDM2 dissociation and nuclear export of p53. Nat Cell Biol. 2007;9:428–35.

    Article  CAS  PubMed  Google Scholar 

  116. Naidu SR, Lakhter AJ, Androphy EJ. PIASy-mediated Tip60 sumoylation regulates p53-induced autophagy. Cell Cycle. 2012;11:2717–28.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  117. Hay RT. SUMO: a history of modification. Mol Cell. 2005;18:1–12.

    Article  CAS  PubMed  Google Scholar 

  118. Wu SY, Chiang CM. Crosstalk between sumoylation and acetylation regulates p53-dependent chromatin transcription and DNA binding. EMBO J. 2009;28:1246–59.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  119. Xirodimas DP, Saville MK, Bourdon JC, Hay RT, Lane DP. Mdm2-mediated NEDD8 conjugation of p53 inhibits its transcriptional activity. Cell. 2004;118:83–97.

    Article  CAS  PubMed  Google Scholar 

  120. Abida WM, Nikolaev A, Zhao W, Zhang W, Gu W. FBXO11 promotes the Neddylation of p53 and inhibits its transcriptional activity. J Biol Chem. 2007;282:1797–804.

    Article  CAS  PubMed  Google Scholar 

  121. Quinn JJ, Chang HY. Unique features of long non-coding RNA biogenesis and function. Nat Rev Genet. 2016;17:47–62.

    Article  CAS  PubMed  Google Scholar 

  122. Statello L, Guo CJ, Chen LL, Huarte M. Author correction: Gene regulation by long non-coding RNAs and its biological functions. Nat Rev Mol Cell Biol. 2021;22:159.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  123. Zhang A, Xu M, Mo YY. Role of the lncRNA-p53 regulatory network in cancer. J Mol Cell Biol. 2014;6:181–91.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  124. Chaudhary R, Lal A. Long noncoding RNAs in the p53 network. Wiley Interdiscip Rev RNA. 2017;8: https://doi.org/10.1002/wrna.1410.

  125. Lin T, Hou PF, Meng S, Chen F, Jiang T, Li ML, et al. Emerging roles of p53 related lncRNAs in cancer progression: a systematic review. Int J Biol Sci. 2019;15:1287–98.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  126. Chen R, Liu Y, Zhuang H, Yang B, Hei K, Xiao M, et al. Corrigendum to article “Quantitative proteomics reveals that long non-coding RNA MALAT1 interacts with DBC1 to regulate p53 acetylation”. Nucleic Acids Res. 2021;49:4199–202.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  127. Ma XY, Wang JH, Wang JL, Ma CX, Wang XC, Liu FS. Malat1 as an evolutionarily conserved lncRNA, plays a positive role in regulating proliferation and maintaining undifferentiated status of early-stage hematopoietic cells. BMC Genomics. 2015;16:676.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  128. Jiang L, Zawacka-Pankau J. The p53/MDM2/MDMX-targeted therapies-a clinical synopsis. Cell Death Dis. 2020;11:237.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  129. Amodio N, Raimondi L, Juli G, Stamato MA, Caracciolo D, Tagliaferri P, et al. MALAT1: a druggable long non-coding RNA for targeted anti-cancer approaches. J Hematol Oncol. 2018;11:63.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  130. Slack FJ, Chinnaiyan AM. The role of non-coding RNAs in oncology. Cell. 2019;179:1033–55.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  131. Jiang MC, Ni JJ, Cui WY, Wang BY, Zhuo W. Emerging roles of lncRNA in cancer and therapeutic opportunities. Am J Cancer Res. 2019;9:1354–66.

    CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

This work was supported by the National Cancer Institute of the National Institutes of Health under Award R35CA253059, RO1CA258390 and R01CA254970 to WG. The content is solely the responsibility of the authors and does not necessarily represent the official views of the National Institutes of Health.

Author information

Authors and Affiliations

Authors

Contributions

ZX wrote and edited the manuscript and prepared the figures. NK, APG and OT proofread and edited the manuscript and the figures. WG conceived and edited the manuscript. All authors approved the final version.

Corresponding author

Correspondence to Wei Gu.

Ethics declarations

Competing interests

OT is currently an employee of AstraZeneca and has stock ownership in AstraZeneca. All other authors declare that they have no conflict of interest.

Additional information

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

Supplementary information

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Xia, Z., Kon, N., Gu, A.P. et al. Deciphering the acetylation code of p53 in transcription regulation and tumor suppression. Oncogene 41, 3039–3050 (2022). https://doi.org/10.1038/s41388-022-02331-9

Download citation

  • Received:

  • Revised:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41388-022-02331-9

This article is cited by

Search

Quick links