Review
Regulation of the p53-MDM2 pathway by 14-3-3 σ and other proteins

https://doi.org/10.1016/j.semcancer.2006.03.009Get rights and content

Abstract

The 14-3-3 σ (sigma) protein, a unique member of 14-3-3 family, is a negative regulator of the cell cycle and is induced by p53 to initiate cell cycle checkpoint control after DNA damage. Among the 14-3-3 family members, 14-3-3 σ is uniquely induced by p53 and has a positive feedback effect on p53 activity in response to DNA damage. Although 14-3-3 σ is linked to p53-regulated cell cycle checkpoint control, the detailed mechanisms of cell cycle regulation by 14-3-3 σ remain unclear. Decreased expression of 14-3-3 σ was reported in several types of carcinomas, suggesting that the negative regulatory role of 14-3-3 σ in the cell cycle is compromised during tumorigenesis. Given the fact that p53's tumor suppressive function is lost in almost half of all human cancers and that 14-3-3 σ's activity is linked to the p53 network, a perspective regarding the p53/14-3-3 σ relationship is needed for cancer research. Here we discuss the mechanisms by which 14-3-3 σ-stabilizes p53 with the hope that these insights may be applied to develop targeted therapeutic strategies for cancer treatment.

Introduction

The 14-3-3 family of proteins has many diverse functions, including critical roles in signal transduction pathways and cell cycle regulation [1], [2], [3]. In mammalian species the 14-3-3 family is comprised of the seven highly conserved isoforms β, ɛ, η, γ, τ (also called θ), ζ, and σ. Among the family members, 14-3-3 σ is unique. The 14-3-3 σ gene is induced by the p53 tumor suppressor protein in response to DNA damage and differs from other isoforms in structure. 14-3-3 σ has unique amino acids (Met202, Asp204, and His206) that may be responsible for binding to particular ligands that are not recognized by other 14-3-3 members [4], [5].

14-3-3 σ was originally characterized as a human mammary epithelial-specific marker (HME1) [6] that is down-regulated in mammary carcinoma cells. 14-3-3 σ negatively regulates the cell cycle by interacting with cylin-dependent kinases (CDKs) [7]. 14-3-3 σ shares cyclin–Cdk2 binding motifs with different cell cycle regulators, including p107, p130, p21Cip1, p27Kip1, and p57Kip2 and is associated with cyclin–Cdk complexes in vitro and in vivo. Reintroduction of 14-3-3 σ in breast cancer cell lines leads to inhibition of CDK-associated histone H1 kinase activity and inhibition of cell growth. After DNA damage 14-3-3 σ is induced by p53. Immunofluorescence studies indicate that 14-3-3 σ sequesters Cdc2 and Cdk2 complexes from the nucleus to the cytoplasm, which contributes to the G2 arrest in response to DNA damage [7], [8]. Importantly, 14-3-3 σ also positively regulates p53 stability and potentiates p53 transcriptional activity [9], thereby executing a positive feedback effect on p53 activity.

Given 14-3-3 σ's negative role in cell growth and positive role in potentiating p53 activity, it is conceivable that 14-3-3 σ plays an important role in controlling cancer formation. Indeed, overexpression of 14-3-3 σ can antagonize oncogene-mediated cell growth and transformation in breast cancer cell lines [7]. Conversely, experimental down regulation of 14-3-3 σ allows primary human epithelial cells to grow indefinitely [10], suggesting that a decrease in 14-3-3 σ expression may contribute to tumor formation by promoting cellular immortalization. Importantly, 14-3-3 σ is downregulated in several types of cancer, including breast cancer [11], [12], ovarian cancer [13], salivary gland adenoid cystic carcinoma [14], gastric cancer [15], hepatocellular carcinoma [16], prostate cancer [17], [18], [19], basal cell carcinoma [20], and lung cancer [21]. Usually, the downregulation of 14-3-3 σ is the result of epigenetic silencing by CpG methylation rather than genetic alteration [22], [23]. These observations suggest that the tumor suppressor function of 14-3-3 σ is compromised during tumorigenesis. Recent data indicate that 14-3-3 σ is efficient in inhibiting the tumorigenicity of ErbB2-overexpressing cells [9], Akt-activating cells [24], [25] and nasopharyngeal carcinoma cells [26]. These data provide compelling evidence that 14-3-3 σ can function as a tumor suppressor.

The observation that 14-3-3 σ has positive feedback effect on p53 activity after DNA damage is very intriguing. However, the mechanisms of 14-3-3 σ's role in p53 stabilization and signal transduction have not been fully elucidated. 14-3-3 σ may regulate potential target proteins involved in p53 stabilization by sub-compartimental sequestration, modulating enzyme activity, and facilitating protein–protein interaction. Here we review the potential mechanisms that may be involved in 14-3-3 σ's positive impact on p53 stabilization with a hope that these insights can be applied to investigate 14-3-3 σ's function in the DNA damage response and tumor suppression.

Section snippets

14-3-3 σ is induced by p53 in response to DNA damage

Importantly, 14-3-3 σ is the only 14-3-3 isoform induced by tumor suppressor protein p53 in response to gamma irradiation and other DNA-damaging agents [27]. The 14-3-3 σ promoter contains a p53 response element; therefore, p53 can directly transactivate the expression of 14-3-3 σ. 14-3-3 σ induction results in a G2 arrest [27]. p53, dubbed ‘a guardian of the genome’ [28], is a critical regulator of cell proliferation in response to DNA damage. p53 activation causes cell arrest, and subsequent

14-3-3 σ has a positive feedback effect on p53

The 14-3-3 σ gene was identified as a p53-inducible gene involved in cell cycle checkpoint control after DNA damage [27]. The 14-3-3 σ protein was also characterized as a negative regulator of cyclin-dependent kinases (CDKs) [7]. Thus, the negative roles of p53 and 14-3-3 σ in cell cycle progression suggested a possible linkage of p53 and 14-3-3 σ. Members of the 14-3-3 protein family have been shown to act as adapter proteins binding to many signal proteins to exert biological function [34].

Regulation of p53 ubiquitin ligases as a mechanism leading to 14-3-3 σ-mediated p53 stabilization

As indicated above, increased levels of 14-3-3 σ leads to decreasing MDM2 stability thus antagonizing MDM2 activity [9]. Because MDM2 mediates its own ubiquitination and subsequent destabilization in a RING finger-dependent manner, it is possible that 14-3-3 σ affects MDM2 self-ubiquitination. In addition, because 14-3-3 σ has a negative impact on MDM2, it is possible that MDM2-mediated modification on p53 could be diminished. For example, one possible regulation is the neddylation process.

Regulation of MDM2 regulators and 14-3-3 σ-mediated p53 stabilization

To further explore how 14-3-3 σ negatively regulates the activity of MDM2 and thereby stabilizes p53, we will review some of the important mechanisms by which MDM2 activity is regulated (Fig. 3). The data presented thus far clearly indicate that 14-3-3 σ negatively regulates MDM2 and subsequently potentiates the activity of p53. As the loss of 14-3-3 σ occurs in many tumors, the presumably resulting increase in levels of MDM2 may inhibit p53 activity. Numerous other proteins however also

Summary

In this review, we have discussed several modes of MDM2 regulation. The potential involvement of 14-3-3 σ in negatively regulating MDM2 activity could be linked to these regulatory mechanisms. Thus, all the discussed proteins are subject to regulation by 14-3-3 σ. For example, 14-3-3 σ may be able to potentiate the activity of MDM2 negative regulators (Fig. 3, Fig. 4), including ARF, L5, L11, L23, but antagonize the MDM2 positive regulators, including Gankyrin, YY1, KAP1 (Fig. 3, Fig. 4), to

Acknowledgements

We would like to thank the William McGowan Charitable Foundation, the Susan Komen Breast Cancer Foundation, and NIH grant (RO1CA 089266 to M.H. Lee, CA47296 to G. Lozano) for research support. We apologize to our many colleagues whose work that we were unable to cite due to space constraints.

References (98)

  • E.S. Stavridi et al.

    Crystal structure of the FHA domain of the Chfr mitotic checkpoint protein and its complex with tungstate

    Structure (Camb)

    (2002)
  • M. Gostissa et al.

    The transcriptional repressor hDaxx potentiates p53-dependent apoptosis

    J Biol Chem

    (2004)
  • D.P. Xirodimas et al.

    Mdm2-mediated NEDD8 conjugation of p53 inhibits its transcriptional activity

    Cell

    (2004)
  • R.P. Leng et al.

    Pirh2, a p53-induced ubiquitin–protein ligase, promotes p53 degradation

    Cell

    (2003)
  • I.R. Logan et al.

    Control of human PIRH2 protein stability: involvement of TIP60 and the proteosome

    J Biol Chem

    (2004)
  • E. Bianchi et al.

    Characterization of human constitutive photomorphogenesis protein 1, a RING finger ubiquitin ligase that interacts with Jun transcription factors and modulates their transcriptional activity

    J Biol Chem

    (2003)
  • S.R. Datta et al.

    14-3-3 proteins and survival kinases cooperate to inactivate BAD by BH3 domain phosphorylation

    Mol Cell

    (2000)
  • A. Brunet et al.

    Akt promotes cell survival by phosphorylating and inhibiting a Forkhead transcription factor

    Cell

    (1999)
  • Y. Ogawara et al.

    Akt enhances Mdm2-mediated ubiquitination and degradation of p53

    J Biol Chem

    (2002)
  • J. Feng et al.

    Stabilization of Mdm2 via decreased ubiquitination is mediated by protein kinase B/Akt-dependent phosphorylation

    J Biol Chem

    (2004)
  • D.J. Freeman et al.

    PTEN tumor suppressor regulates p53 protein levels and activity through phosphatase-dependent and -independent mechanisms

    Cancer Cell

    (2003)
  • V. Stambolic et al.

    Regulation of PTEN transcription by p53

    Mol Cell

    (2001)
  • J. Puc et al.

    Lack of PTEN sequesters CHK1 and initiates genetic instability

    Cancer Cell

    (2005)
  • T. Kamijo et al.

    Tumor suppression at the mouse INK4a locus mediated by the alternative reading frame product p19ARF

    Cell

    (1997)
  • D. Chen et al.

    ARF-BP1/Mule is a critical mediator of the ARF tumor suppressor

    Cell

    (2005)
  • S. Adhikary et al.

    The ubiquitin ligase HectH9 regulates transcriptional activation by Myc and is essential for tumor cell proliferation

    Cell

    (2005)
  • O. Vafa et al.

    c-Myc can induce DNA damage, increase reactive oxygen species, and mitigate p53 function: a mechanism for oncogene-induced genetic instability

    Mol Cell

    (2002)
  • M.S. Dai et al.

    Inhibition of MDM2-mediated p53 ubiquitination and degradation by ribosomal protein L5

    J Biol Chem

    (2004)
  • M.A. Lohrum et al.

    Regulation of HDM2 activity by the ribosomal protein L11

    Cancer Cell

    (2003)
  • B.J. Park et al.

    The haploinsufficient tumor suppressor p18 upregulates p53 via interactions with ATM/ATR

    Cell

    (2005)
  • H. Higashitsuji et al.

    The oncoprotein gankyrin binds to MDM2/HDM2, enhancing ubiquitylation and degradation of p53

    Cancer Cell

    (2005)
  • M. Li et al.

    A dynamic role of HAUSP in the p53-Mdm2 pathway

    Mol Cell

    (2004)
  • E. Meulmeester et al.

    Loss of HAUSP-mediated deubiquitination contributes to DNA damage-induced destabilization of Hdmx and Hdm2

    Mol Cell

    (2005)
  • G. Sui et al.

    Yin Yang 1 is a negative regulator of p53

    Cell

    (2004)
  • H. Vaziri et al.

    hSIR2(SIRT1) functions as an NAD-dependent p53 deacetylase

    Cell

    (2001)
  • T. Kahyo et al.

    Involvement of PIAS1 in the sumoylation of tumor suppressor p53

    Mol Cell

    (2001)
  • H. Fu et al.

    14-3-3 proteins: structure, function, and regulation

    Annu Rev Pharmacol Toxicol

    (2000)
  • A. Benzinger et al.

    The crystal structure of the non-liganded 14-3-3sigma protein: insights into determinants of isoform specific ligand binding and dimerization

    Cell Res

    (2005)
  • G.L. Prasad et al.

    Complementary DNA cloning of a novel epithelial cell marker protein, Hme1, that may be down-regulated in neoplastic mammary cells

    Cell Growth Differ

    (1992)
  • T.A. Chan et al.

    14-3-3sigma is required to prevent mitotic catastrophe after DNA damage

    Nature

    (1999)
  • H.Y. Yang et al.

    14-3-3sigma positively regulates p53 and suppresses tumor growth

    Mol Cell Biol

    (2003)
  • E. Dellambra et al.

    Downregulation of 14-3-3sigma prevents clonal evolution and leads to immortalization of primary human keratinocytes

    J Cell Biol

    (2000)
  • A.T. Ferguson et al.

    High frequency of hypermethylation at the 14-3-3 sigma locus leads to gene silencing in breast cancer

    Proc Natl Acad Sci USA

    (2000)
  • J. Akahira et al.

    Decreased expression of 14-3-3 sigma is associated with advanced disease in human epithelial ovarian cancer: its correlation with aberrant DNA methylation

    Clin Cancer Res

    (2004)
  • D. Uchida et al.

    Frequent downregulation of 14-3-3 sigma protein and hypermethylation of 14-3-3 sigma gene in salivary gland adenoid cystic carcinoma

    Br J Cancer

    (2004)
  • H. Suzuki et al.

    Inactivation of the 14-3-3 sigma gene is associated with 5′ CpG island hypermethylation in human cancers

    Cancer Res

    (2000)
  • N. Iwata et al.

    Frequent hypermethylation of CpG islands and loss of expression of the 14-3-3 sigma gene in human hepatocellular carcinoma

    Oncogene

    (2000)
  • D. Lodygin et al.

    Prostate cancer is characterized by epigenetic silencing of 14-3-3sigma expression

    Oncogene

    (2004)
  • R. Henrique et al.

    Frequent 14-3-3 sigma promoter methylation in benign and malignant prostate lesions

    DNA Cell Biol

    (2005)
  • Cited by (93)

    • 14-3-3 σ: A potential biomolecule for cancer therapy

      2020, Clinica Chimica Acta
      Citation Excerpt :

      After being induced by γ-ray or DNA-damaging drugs, p53 transactivates the expression of 14-3-3σ, resulting in cell cycle arrest at G2/M checkpoints by sequestering CDK2/cyclin B complexes. The existence of 14-3-3σ is mainly to block the cell cycle at the G2-M checkpoint [38,39]. In contrast, HCT116 cell lines lacking 14-3-3σ entered mitotic disaster after DNA-damaging drug treatment [40].

    • Recent advances in the development of ubiquitin-specific-processing protease 7 (USP7) inhibitors

      2020, European Journal of Medicinal Chemistry
      Citation Excerpt :

      Abnormal expression of USP7 not only triggers mental diseases [41], but also chemo- and radio-resistance [42], owing to the multiple roles of USP7. It is specifically involved in multi-intracellular processes, such as DNA damage (Claspin, CHFR), immune response (TRAF6, IKKγ), tumour suppression (HDM2, FOXO4, HDMX, PTEN), viral infection (ICP0, EBNA1) and epigenetic control (MEL18, BMI1, H2B) [43–46]. Probably the reactive oxygen species reversibly inactivate USP7 by oxidation of the catalytic cysteine residue, thus triggering an abnormal USP7 function, which further affects other cellular pathways as response to oxidative stress [47].

    • 14-3-3 protein sigma isoform co-localizes with phosphorylated α-synuclein in Lewy bodies and Lewy neurites in patients with Lewy body disease

      2018, Neuroscience Letters
      Citation Excerpt :

      In the presence of rotenone, 14-3-3 sigma cells showed increased toxicity compared to control cells. The sigma isoform has potential not only for cell cycle control, but also for p53 regulation [12]; p53 involvement in PD pathology has been reported [6]. Satoh et al. [17] suggested that the sigma isoform might serve as a marker of oxidative and DNA-damaging stresses inducing the mitotic checkpoint dysfunction in reactive astrocytes under pathological conditions.

    View all citing articles on Scopus
    View full text