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.

  • Original Article
  • Published:

The pro-longevity gene FoxO3 is a direct target of the p53 tumor suppressor

Abstract

FoxO transcription factors have a conserved role in longevity, and act as tissue-specific tumor suppressors in mammals. Several nodes of interaction have been identified between FoxO transcription factors and p53, a major tumor suppressor in humans and mice. However, the extent and importance of the functional interaction between FoxO and p53 have not been fully explored. Here, we show that p53 regulates the expression of FoxO3, one of the four mammalian FoxO genes, in response to DNA damaging agents in both mouse embryonic fibroblasts and thymocytes. We find that p53 transactivates FoxO3 in cells by binding to a site in the second intron of the FoxO3 gene, a genomic region recently found to be associated with extreme longevity in humans. While FoxO3 is not necessary for p53-dependent cell cycle arrest, FoxO3 appears to modulate p53-dependent apoptosis. We also find that FoxO3 loss does not interact with p53 loss for tumor development in vivo, although the tumor spectrum of p53-deficient mice appears to be affected by FoxO3 loss. Our findings indicate that FoxO3 is a p53 target gene, and suggest that FoxO3 and p53 are part of a regulatory transcriptional network that may have an important role during aging and cancer.

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

Figure 1
Figure 2
Figure 3
Figure 4
Figure 5
Figure 6
Figure 7
Figure 8

Similar content being viewed by others

References

  • Anselmi CV, Malovini A, Roncarati R, Novelli V, Villa F, Condorelli G et al. (2009). Association of the FOXO3A locus with extreme longevity in a southern Italian centenarian study. Rejuvenation Res 12: 95–104.

    CAS  PubMed  Google Scholar 

  • Arum O, Johnson TE . (2007). Reduced expression of the Caenorhabditis elegans p53 ortholog cep-1 results in increased longevity. J Gerontol A Biol Sci Med Sci 62: 951–959.

    PubMed  Google Scholar 

  • Attardi LD, Reczek EE, Cosmas C, Demicco EG, McCurrach ME, Lowe SW et al. (2000). PERP, an apoptosis-associated target of p53, is a novel member of the PMP-22/gas3 family. Genes Dev 14: 704–718.

    CAS  PubMed  PubMed Central  Google Scholar 

  • Bakker WJ, Harris IS, Mak TW . (2007). FOXO3a is activated in response to hypoxic stress and inhibits HIF1-induced apoptosis via regulation of CITED2. Mol Cell 28: 941–953.

    CAS  PubMed  Google Scholar 

  • Bauer JH, Poon PC, Glatt-Deeley H, Abrams JM, Helfand SL . (2005). Neuronal expression of p53 dominant-negative proteins in adult Drosophila melanogaster extends life span. Curr Biol 15: 2063–2068.

    CAS  PubMed  Google Scholar 

  • Bluher M, Kahn BB, Kahn CR . (2003). Extended longevity in mice lacking the insulin receptor in adipose tissue. Science 299: 572–574.

    PubMed  Google Scholar 

  • Bouchard C, Lee S, Paulus-Hock V, Loddenkemper C, Eilers M, Schmitt CA . (2007). FoxO transcription factors suppress Myc-driven lymphomagenesis via direct activation of Arf. Genes Dev 21: 2775–2787.

    CAS  PubMed  PubMed Central  Google Scholar 

  • Brunet A, Bonni A, Zigmond MJ, Lin MZ, Juo P, Hu LS et al. (1999). Akt promotes cell survival by phosphorylating and inhibiting a Forkhead transcription factor. Cell 96: 857–868.

    CAS  PubMed  Google Scholar 

  • Brunet A, Datta SR, Greenberg ME . (2001). Transcription-dependent and -independent control of neuronal survival by the PI3K-Akt signaling pathway. Curr Opin Neurobiol 11: 297–305.

    CAS  PubMed  Google Scholar 

  • Brunet A, Kanai F, Stehn J, Xu J, Sarbassova D, Frangioni JV et al. (2002). 14–3–3 transits to the nucleus and participates in dynamic nucleocytoplasmic transport. J Cell Biol 156: 817–828.

    CAS  PubMed  PubMed Central  Google Scholar 

  • Brunet A, Sweeney LB, Sturgill JF, Chua KF, Greer PL, Lin Y et al. (2004). Stress-dependent regulation of FOXO transcription factors by the SIRT1 deacetylase. Science 303: 2011–2015.

    CAS  PubMed  Google Scholar 

  • Calnan DR, Brunet A . (2008). The FoxO code. Oncogene 27: 2276–2288.

    CAS  PubMed  Google Scholar 

  • Castrillon DH, Miao L, Kollipara R, Horner JW, DePinho RA . (2003). Suppression of ovarian follicle activation in mice by the transcription factor Foxo3a. Science 301: 215–218.

    CAS  PubMed  Google Scholar 

  • de la Torre-Ubieta L, Gaudilliere B, Yang Y, Ikeuchi Y, Yamada T, DiBacco S et al. (2010). A FOXO-Pak1 transcriptional pathway controls neuronal polarity. Genes Dev 24: 799–813.

    CAS  PubMed  PubMed Central  Google Scholar 

  • Deng G, Chen LC, Schott DR, Thor A, Bhargava V, Ljung BM et al. (1994). Loss of heterozygosity and p53 gene mutations in breast cancer. Cancer Res 54: 499–505.

    CAS  PubMed  Google Scholar 

  • Dijkers PF, Medemadagger RH, Lammers JJ, Koenderman L, Coffer PJ . (2000). Expression of the pro-apoptotic bcl-2 family member bim is regulated by the forkhead transcription factor FKHR-L1. Curr Biol 10: 1201–1204.

    CAS  PubMed  Google Scholar 

  • Donehower LA, Harvey M, Slagle BL, McArthur MJ, Montgomery Jr CA, Butel JS et al. (1992). Mice deficient for p53 are developmentally normal but susceptible to spontaneous tumours. Nature 356: 215–221.

    CAS  PubMed  Google Scholar 

  • el-Deiry WS, Kern SE, Pietenpol JA, Kinzler KW, Vogelstein B . (1992). Definition of a consensus binding site for p53. Nat Genet 1: 45–49.

    CAS  PubMed  Google Scholar 

  • el-Deiry WS, Tokino T, Velculescu VE, Levy DB, Parsons R, Trent JM et al. (1993). WAF1, a potential mediator of p53 tumor suppression. Cell 75: 817–825.

    CAS  PubMed  Google Scholar 

  • Essaghir A, Dif N, Marbehant CY, Coffer PJ, Demoulin JB . (2009). The transcription of FOXO genes is stimulated by FOXO3 and repressed by growth factors. J Biol Chem 284: 10334–10342.

    CAS  PubMed  PubMed Central  Google Scholar 

  • Fiscella M, Zhang H, Fan S, Sakaguchi K, Shen S, Mercer WE et al. (1997). Wip1, a novel human protein phosphatase that is induced in response to ionizing radiation in a p53-dependent manner. Proc Natl Acad Sci USA 94: 6048–6053.

    CAS  PubMed  PubMed Central  Google Scholar 

  • Flachsbart F, Caliebe A, Kleindorp R, Blanche H, von Eller-Eberstein H, Nikolaus S et al. (2009). Association of FOXO3A variation with human longevity confirmed in German centenarians. Proc Natl Acad Sci USA 106: 2700–2705.

    CAS  PubMed  PubMed Central  Google Scholar 

  • Fu W, Ma Q, Chen L, Li P, Zhang M, Ramamoorthy S et al. (2009). MDM2 acts downstream of p53 as an E3 ligase to promote FOXO ubiquitination and degradation. J Biol Chem 284: 13987–14000.

    CAS  PubMed  PubMed Central  Google Scholar 

  • Furuyama T, Yamashita H, Kitayama K, Higami Y, Shimokawa I, Mori N . (2002). Effects of aging and caloric restriction on the gene expression of Foxo1, 3, and 4 (FKHR, FKHRL1, and AFX) in the rat skeletal muscles. Microsc Res Tech 59: 331–334.

    CAS  PubMed  Google Scholar 

  • Gaidano G, Ballerini P, Gong JZ, Inghirami G, Neri A, Newcomb EW et al. (1991). p53 mutations in human lymphoid malignancies: association with Burkitt lymphoma and chronic lymphocytic leukemia. Proc Natl Acad Sci USA 88: 5413–5417.

    CAS  PubMed  PubMed Central  Google Scholar 

  • Giannakou ME, Goss M, Junger MA, Hafen E, Leevers SJ, Partridge L . (2004). Long-lived drosophila with overexpressed dFOXO in adult fat body. Science 305: 361.

    CAS  PubMed  Google Scholar 

  • Greer EL, Brunet A . (2005). FOXO transcription factors at the interface between longevity and tumor suppression. Oncogene 24: 7410–7425.

    Article  CAS  PubMed  Google Scholar 

  • Greer EL, Dowlatshahi D, Banko MR, Villen J, Hoang K, Blanchard D et al. (2007). An AMPK-FOXO pathway mediates longevity induced by a novel method of dietary restriction in C elegans. Curr Biol 17: 1646–1656.

    CAS  PubMed  PubMed Central  Google Scholar 

  • Harvey M, McArthur MJ, Montgomery Jr CA, Bradley A, Donehower LA . (1993a). Genetic background alters the spectrum of tumors that develop in p53-deficient mice. FASEB J 7: 938–943.

    CAS  PubMed  Google Scholar 

  • Harvey M, McArthur MJ, Montgomery Jr CA, Butel JS, Bradley A, Donehower LA . (1993b). Spontaneous and carcinogen-induced tumorigenesis in p53-deficient mice. Nat Genet 5: 225–229.

    CAS  PubMed  Google Scholar 

  • Henderson ST, Johnson TE . (2001). daf-16 integrates developmental and environmental inputs to mediate aging in the nematode Caenorhabditis elegans. Curr Biol 11: 1975–1980.

    CAS  PubMed  Google Scholar 

  • Holzenberger M, Dupont J, Ducos B, Leneuve P, Geloen A, Even PC et al. (2003). IGF-1 receptor regulates lifespan and resistance to oxidative stress in mice. Nature 421: 182–187.

    CAS  PubMed  Google Scholar 

  • Hribal ML, Nakae J, Kitamura T, Shutter JR, Accili D . (2003). Regulation of insulin-like growth factor-dependent myoblast differentiation by Foxo forkhead transcription factors. J Cell Biol 162: 535–541.

    CAS  PubMed  PubMed Central  Google Scholar 

  • Hu MC, Lee DF, Xia W, Golfman LS, Ou-Yang F, Yang JY et al. (2004). IkappaB kinase promotes tumorigenesis through inhibition of forkhead FOXO3a. Cell 117: 225–237.

    CAS  PubMed  Google Scholar 

  • Hursting SD, Lavigne JA, Berrigan D, Perkins SN, Barrett JC . (2003). Calorie restriction, aging, and cancer prevention: mechanisms of action and applicability to humans. Annu Rev Med 54: 131–152.

    CAS  PubMed  Google Scholar 

  • Hwangbo DS, Gersham B, Tu MP, Palmer M, Tatar M . (2004). Drosophila dFOXO controls lifespan and regulates insulin signalling in brain and fat body. Nature 429: 562–566.

    CAS  PubMed  Google Scholar 

  • Ihrie RA, Reczek E, Horner JS, Khachatrian L, Sage J, Jacks T et al. (2003). Perp is a mediator of p53-dependent apoptosis in diverse cell types. Curr Biol 13: 1985–1990.

    CAS  PubMed  Google Scholar 

  • Imae M, Fu Z, Yoshida A, Noguchi T, Kato H . (2003). Nutritional and hormonal factors control the gene expression of FoxOs, the mammalian homologues of DAF-16. J Mol Endocrinol 30: 253–262.

    CAS  PubMed  Google Scholar 

  • Jacks T, Remington L, Williams BO, Schmitt EM, Halachmi S, Bronson RT et al. (1994). Tumor spectrum analysis in p53-mutant mice. Curr Biol 4: 1–7.

    CAS  PubMed  Google Scholar 

  • Jacobs SB, Basak S, Murray JI, Pathak N, Attardi LD . (2007). Siva is an apoptosis-selective p53 target gene important for neuronal cell death. Cell Death Differ 14: 1374–1385.

    CAS  PubMed  Google Scholar 

  • Johnson TM, Hammond EM, Giaccia A, Attardi LD . (2005). The p53QS transactivation-deficient mutant shows stress-specific apoptotic activity and induces embryonic lethality. Nat Genet 37: 145–152.

    CAS  PubMed  Google Scholar 

  • Johnson TM, Meade K, Pathak N, Marques MR, Attardi LD . (2008). Knockin mice expressing a chimeric p53 protein reveal mechanistic differences in how p53 triggers apoptosis and senescence. Proc Natl Acad Sci USA 105: 1215–1220.

    CAS  PubMed  PubMed Central  Google Scholar 

  • Juven T, Barak Y, Zauberman A, George DL, Oren M . (1993). Wild type p53 can mediate sequence-specific transactivation of an internal promoter within the mdm2 gene. Oncogene 8: 3411–3416.

    CAS  PubMed  Google Scholar 

  • Kastan MB, Zhan Q, el-Deiry WS, Carrier F, Jacks T, Walsh WV et al. (1992). A mammalian cell cycle checkpoint pathway utilizing p53 and GADD45 is defective in ataxia-telangiectasia. Cell 71: 587–597.

    CAS  PubMed  Google Scholar 

  • Kenyon C . (2005). The plasticity of aging: insights from long-lived mutants. Cell 120: 449–460.

    CAS  PubMed  Google Scholar 

  • Kops GJ, Dansen TB, Polderman PE, Saarloos I, Wirtz KW, Coffer PJ et al. (2002). Forkhead transcription factor FOXO3a protects quiescent cells from oxidative stress. Nature 419: 316–321.

    CAS  PubMed  Google Scholar 

  • Kurinna S, Stratton SA, Tsai WW, Akdemir KC, Gu W, Singh P et al. (2010). Direct activation of forkhead box O3 by tumor suppressors p53 and p73 is disrupted during liver regeneration in mice. Hepatology 52: 1023–1032.

    CAS  PubMed  Google Scholar 

  • Lenormand P, Sardet C, Pagès G, L'Allemain G, Brunet A, Pouysségur J . (1993). Growth factors induce nuclear translocation of MAP kinases (p42mapk and p44mapk) but not of their activator MAP kinase kinase (p45 mapkk) in fibroblasts. J Cell Biol 122: 1079–1088.

    CAS  PubMed  Google Scholar 

  • Lo Coco F, Gaidano G, Louie DC, Offit K, Chaganti RS, Dalla-Favera R . (1993). p53 mutations are associated with histologic transformation of follicular lymphoma. Blood 82: 2289–2295.

    CAS  PubMed  Google Scholar 

  • Lowe SW, Ruley HE . (1993). Stabilization of the p53 tumor suppressor is induced by adenovirus 5 E1A and accompanies apoptosis. Genes Dev 7: 535–545.

    CAS  PubMed  Google Scholar 

  • Lowe SW, Ruley HE, Jacks T, Housman DE . (1993). p53-dependent apoptosis modulates the cytotoxicity of anticancer agents. Cell 74: 957–967.

    CAS  PubMed  Google Scholar 

  • Luo J, Nikolaev AY, Imai S, Chen D, Su F, Shiloh A et al. (2001). Negative control of p53 by Sir2alpha promotes cell survival under stress. Cell 107: 137–148.

    CAS  PubMed  Google Scholar 

  • Maier B, Gluba W, Bernier B, Turner T, Mohammad K, Guise T et al. (2004). Modulation of mammalian life span by the short isoform of p53. Genes Dev 18: 306–319.

    CAS  PubMed  PubMed Central  Google Scholar 

  • Marusyk A, Wheeler LJ, Mathews CK, DeGregori J . (2007). p53 mediates senescence-like arrest induced by chronic replicational stress. Mol Cell Biol 27: 5336–5351.

    CAS  PubMed  PubMed Central  Google Scholar 

  • Masoro EJ . (2005). Overview of caloric restriction and ageing. Mech Ageing Dev 126: 913–922.

    CAS  PubMed  Google Scholar 

  • Matheu A, Maraver A, Klatt P, Flores I, Garcia-Cao I, Borras C et al. (2007). Delayed ageing through damage protection by the Arf/p53 pathway. Nature 448: 375–379.

    CAS  PubMed  Google Scholar 

  • Medema RH, Kops GJ, Bos JL, Burgering BM . (2000). AFX-like Forkhead transcription factors mediate cell-cycle regulation by Ras and PKB through p27kip1. Nature 404: 782–787.

    CAS  PubMed  Google Scholar 

  • Miyaguchi Y, Tsuchiya K, Sakamoto K . (2009). P53 negatively regulates the transcriptional activity of FOXO3a under oxidative stress. Cell Biol Int 33: 853–860.

    CAS  PubMed  Google Scholar 

  • Motta MC, Divecha N, Lemieux M, Kamel C, Chen D, Gu W et al. (2004). Mammalian SIRT1 represses forkhead transcription factors. Cell 116: 551–563.

    CAS  PubMed  Google Scholar 

  • Nakano K, Vousden KH . (2001). PUMA, a novel proapoptotic gene, is induced by p53. Mol Cell 7: 683–694.

    CAS  PubMed  Google Scholar 

  • Nemoto S, Fergusson MM, Finkel T . (2004). Nutrient availability regulates SIRT1 through a forkhead-dependent pathway. Science 306: 2105–2108.

    CAS  PubMed  Google Scholar 

  • Nowak K, Killmer K, Gessner C, Lutz W . (2007). E2F-1 regulates expression of FOXO1 and FOXO3a. Biochim Biophys Acta 1769: 244–252.

    CAS  PubMed  Google Scholar 

  • Paik JH, Ding Z, Narurkar R, Ramkissoon S, Muller F, Kamoun WS et al. (2009). FoxOs cooperatively regulate diverse pathways governing neural stem cell homeostasis. Cell Stem Cell 5: 540–553.

    CAS  PubMed  PubMed Central  Google Scholar 

  • Paik JH, Kollipara R, Chu G, Ji H, Xiao Y, Ding Z et al. (2007). FoxOs are lineage-restricted redundant tumor suppressors and regulate endothelial cell homeostasis. Cell 128: 309–323.

    CAS  PubMed  PubMed Central  Google Scholar 

  • Pawlikowska L, Hu D, Huntsman S, Sung A, Chu C, Chen J et al. (2009). Association of common genetic variation in the insulin/IGF1 signaling pathway with human longevity. Aging Cell 8: 460–472.

    CAS  PubMed  Google Scholar 

  • Quinlan DC, Davidson AG, Summers CL, Warden HE, Doshi HM . (1992). Accumulation of p53 protein correlates with a poor prognosis in human lung cancer. Cancer Res 52: 4828–4831.

    CAS  PubMed  Google Scholar 

  • Renault VM, Rfafalski VA, Morgan AA, Salih DA, Brett JO, Webb AE et al. (2009). FoxO3 regulates neural stem cell homeostasis. Cell Stem Cell 5: 527–539.

    CAS  PubMed  PubMed Central  Google Scholar 

  • Riley T, Sontag E, Chen P, Levine A . (2008). Transcriptional control of human p53-regulated genes. Nat Rev Mol Cell Biol 9: 402–412.

    CAS  PubMed  Google Scholar 

  • Scott N, Sagar P, Stewart J, Blair GE, Dixon MF, Quirke P . (1991). p53 in colorectal cancer: clinicopathological correlation and prognostic significance. Br J Cancer 63: 317–319.

    CAS  PubMed  PubMed Central  Google Scholar 

  • Seoane J, Le HV, Shen L, Anderson SA, Massague J . (2004). Integration of Smad and forkhead pathways in the control of neuroepithelial and glioblastoma cell proliferation. Cell 117: 211–223.

    CAS  PubMed  Google Scholar 

  • Sharpless NE, Alson S, Chan S, Silver DP, Castrillon DH, DePinho RA . (2002). p16(INK4a) and p53 deficiency cooperate in tumorigenesis. Cancer Res 62: 2761–2765.

    CAS  PubMed  Google Scholar 

  • Shen J, Tower J . (2010). Drosophila foxo acts in males to cause sexual-dimorphism in tissue-specific p53 life span effects. Exp Gerontol 45: 97–105.

    PubMed  Google Scholar 

  • Smith ML, Chen IT, Zhan Q, Bae I, Chen CY, Gilmer TM et al. (1994). Interaction of the p53-regulated protein Gadd45 with proliferating cell nuclear antigen. Science 266: 1376–1380.

    CAS  PubMed  Google Scholar 

  • Takimoto R, El-Deiry WS . (2000). Wild-type p53 transactivates the KILLER/DR5 gene through an intronic sequence-specific DNA-binding site. Oncogene 19: 1735–1743.

    CAS  PubMed  Google Scholar 

  • Tothova Z, Kollipara R, Huntly BJ, Lee BH, Castrillon DH, Cullen DE et al. (2007). FoxOs are critical mediators of hematopoietic stem cell resistance to physiologic oxidative stress. Cell 128: 325–339.

    CAS  PubMed  Google Scholar 

  • Tran H, Brunet A, Grenier JM, Datta SR, Fornace Jr AJ, DiStefano PS et al. (2002). DNA repair pathway stimulated by the forkhead transcription factor FOXO3a through the Gadd45 protein. Science 296: 530–534.

    CAS  PubMed  Google Scholar 

  • Tyner SD, Venkatachalam S, Choi J, Jones S, Ghebranious N, Igelmann H et al. (2002). p53 mutant mice that display early ageing-associated phenotypes. Nature 415: 45–53.

    CAS  PubMed  Google Scholar 

  • Van Der Heide LP, Hoekman MF, Smidt MP . (2004). The ins and outs of FoxO shuttling: mechanisms of FoxO translocation and transcriptional regulation. Biochem J 380 (Part 2): 297–309.

    CAS  PubMed  PubMed Central  Google Scholar 

  • Vassilev LT, Vu BT, Graves B, Carvajal D, Podlaski F, Filipovic Z et al. (2004). in vivo activation of the p53 pathway by small-molecule antagonists of MDM2. Science 303: 844–848.

    CAS  PubMed  Google Scholar 

  • Vaziri H, Dessain SK, Ng Eaton E, Imai SI, Frye RA, Pandita TK et al. (2001). hSIR2(SIRT1) functions as an NAD-dependent p53 deacetylase. Cell 107: 149–159.

    CAS  PubMed  Google Scholar 

  • Velasco-Miguel S, Buckbinder L, Jean P, Gelbert L, Talbott R, Laidlaw J et al. (1999). PA26, a novel target of the p53 tumor suppressor and member of the GADD family of DNA damage and growth arrest inducible genes. Oncogene 18: 127–137.

    CAS  PubMed  Google Scholar 

  • Ventura A, Meissner A, Dillon CP, McManus M, Sharp PA, Van Parijs L et al. (2004). Cre-lox-regulated conditional RNA interference from transgenes. Proc Natl Acad Sci USA 101: 10380–10385.

    CAS  PubMed  PubMed Central  Google Scholar 

  • Vogelstein B, Lane D, Levine AJ . (2000). Surfing the p53 network. Nature 408: 307–310.

    CAS  PubMed  Google Scholar 

  • Vousden KH, Lu X . (2002). Live or let die: the cell's response to p53. Nat Rev Cancer 2: 594–604.

    CAS  PubMed  Google Scholar 

  • Vousden KH, Prives C . (2009). Blinded by the light: the growing complexity of p53. Cell 137: 413–431.

    CAS  PubMed  Google Scholar 

  • Wang F, Marshall CB, Yamamoto K, Li GY, Plevin MJ, You H et al. (2008). Biochemical and structural characterization of an intramolecular interaction in FOXO3a and its binding with p53. J Mol Biol 384: 590–603.

    CAS  PubMed  Google Scholar 

  • Wei CL, Wu Q, Vega VB, Chiu KP, Ng P, Zhang T et al. (2006). A global map of p53 transcription-factor binding sites in the human genome. Cell 124: 207–219.

    CAS  PubMed  Google Scholar 

  • Willcox BJ, Donlon TA, He Q, Chen R, Grove JS, Yano K et al. (2008). FOXO3A genotype is strongly associated with human longevity. Proc Natl Acad Sci USA 105: 13987–13992.

    CAS  PubMed  PubMed Central  Google Scholar 

  • Wu X, Bayle JH, Olson D, Levine AJ . (1993). The p53-mdm-2 autoregulatory feedback loop. Genes Dev 7: 1126–1132.

    CAS  PubMed  Google Scholar 

  • You H, Jang Y, You-Ten AI, Okada H, Liepa J, Wakeham A et al. (2004). p53-dependent inhibition of FKHRL1 in response to DNA damage through protein kinase SGK1. Proc Natl Acad Sci USA 101: 14057–14062.

    CAS  PubMed  PubMed Central  Google Scholar 

  • You H, Pellegrini M, Tsuchihara K, Yamamoto K, Hacker G, Erlacher M et al. (2006a). FOXO3a-dependent regulation of Puma in response to cytokine/growth factor withdrawal. J Exp Med 203: 1657–1663.

    CAS  PubMed  PubMed Central  Google Scholar 

  • You H, Yamamoto K, Mak TW . (2006b). Regulation of transactivation-independent proapoptotic activity of p53 by FOXO3a. Proc Natl Acad Sci USA 103: 9051–9056.

    CAS  PubMed  PubMed Central  Google Scholar 

  • Yu J, Zhang L, Hwang PM, Kinzler KW, Vogelstein B . (2001). PUMA induces the rapid apoptosis of colorectal cancer cells. Mol Cell 7: 673–682.

    CAS  PubMed  Google Scholar 

  • Zhao R, Gish K, Murphy M, Yin Y, Notterman D, Hoffman WH et al. (2000). Analysis of p53-regulated gene expression patterns using oligonucleotide arrays. Genes Dev 14: 981–993.

    CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank Dr Ron DePinho for his generous gift of the FoxO3−/− mice. We thank Julien Sage for critical discussion, reading of the manuscript, help with tumor analysis and for taking the pictures for Figure 8d. We thank Jamie Brett for reading the manuscript. We thank Pauline Chu (Comparative Medicine Department, Stanford Medical School) for her help in processing the histopathology samples. We thank Dan Calnan for participating in earlier aspects of this work. This work was supported by NIH R01 AG026648 and a McCormick Award for Women in Science (AB). ZX, EES and MQZ were supported partly by NIH R01 HG001696. VMR received support from the Dean's Fellowship at Stanford University.

Author contributions: VMR designed, performed and analyzed the lifespan and tumor spectrum of compound FoxO3/p53 mice (Figure 8). PUT completed Figure 1b, Figure 5b, Figure 6b, Figure 6e, Figure 7a and helped with Figure 2 and Figure 6d. KLH completed Figure 5d, Figure 5e, Figure 6a, Figure 6b and Figure 6c. JLW completed Figure 2, Figure 6d, and helped with Figure 3 and Figure 7b. CAB completed Figure 3 and Figure 7b. DKB completed Figure 5c. OSV initiated this project as her Honors Thesis and completed Figure 1a. TMJ completed Figure 4b. PRO completed Figure 6f. ZX and EES identified p53 binding sites in FoxO3 regulatory regions (Figure 5a) under the supervision of MQZ. HV performed the tumor identification (Figure 8b). LDA supervised CAB, DKB and TMJ, and provided ideas. AB supervised VMR, PUT, KLH, JLW, OSV and PRO. The manuscript was written by AB, VMR and OSV, with input from LDA.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to A Brunet.

Ethics declarations

Competing interests

The authors declare no conflict of interest.

Rights and permissions

Reprints and permissions

About this article

Cite this article

Renault, V., Thekkat, P., Hoang, K. et al. The pro-longevity gene FoxO3 is a direct target of the p53 tumor suppressor. Oncogene 30, 3207–3221 (2011). https://doi.org/10.1038/onc.2011.35

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/onc.2011.35

Keywords

This article is cited by

Search

Quick links