p53 suppresses 14-3-3γ by stimulating proteasome-mediated 14-3-3γ protein degradation

  • Authors:
    • De-Yu Chen
    • Dong-Fang Dai
    • Ye Hua
    • Wen-Qing Qi
  • View Affiliations

  • Published online on: November 7, 2014     https://doi.org/10.3892/ijo.2014.2740
  • Pages: 818-824
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

14-3-3 proteins are a family of highly conserved polypeptides that interact with a large number of proteins and play a role in a wide variety of cellular processes. 14-3-3 proteins have been demonstrated overexpressed in several cancers and serving as potential oncogenes. In a previous study we showed one isoform of the 14-3-3 family, 14-3-3γ was negatively regulated by p53 through binding to its promoter and inhibiting its transcription. In the present study we investigated both p53 and 14-3-3γ protein levels in human lung cancerous tissues and normal lung tissues. We found 14-3-3γ expression correlated to p53 overexpression in lung cancer tissues. Ecotopic expression of wild-type p53, but not mutant p53 (R175H) suppressed both endogenous and exogenous 14-3-3γ in colon and lung cancer cell lines. Further examination demonstrated that p53 interacted with C-terminal domain of 14-3-3γ and induced 14-3-3γ ubiquitination. MG132, a specific inhibitor of the 26S proteasome, could block the effect of p53 on 14-3-3γ protein levels, suggesting that p53 suppressed 14-3-3γ by stimulating the process of proteasome-mediated degradation of 14-3-3γ. These results indicate that the inhibitory effect of p53 on 14-3-3γ is mediated also by a post-transcriptional mechanism. Loss of p53 function may result in upregulation of 14-3-3γ in lung cancers.

Introduction

The 14-3-3 proteins are a family of highly conserved acidic polypeptides that are expressed in all eukaryotic cells (13). Seven isoforms, encoded by seven distinct genes have been identified in mammals (named β, γ, ɛ, η, σ, τ and ζ). The 14-3-3 family proteins are thought to act as scaffolding proteins and interact with a wide variety of protein partners which participate in many biological processes, such as cell cycle progression (47), apoptosis (810), cell adhesion and migration (11,12) and signal transduction regulation (1315). Importantly, high expression of 14-3-3 family proteins have been observed in a number of human cancers, including lung (16,17), prostate (18,19), pancreatic (20,21), gastric (22) and colon cancer (23) which suggests that they may have a critical role as tumor oncogenes (24). However, few studies concerning regulation of 14-3-3 protein expression on transcriptional or post-transcriptional levels have been published.

The p53 tumor suppression protein has been defined as a critical component of the DNA damage checkpoint machinery (25). p53 can transcriptionally activate or silence a number of target genes (26,27). One of the seven human 14-3-3 isoforms 14-3-3σ is directly transactivated by p53 after DNA damage (28,29). 14-3-3σ induces cell cycle arrest in G2 by sequestering proteins in the cytoplasm which are required for entry into mitosis (29,30). Noyably, 14-3-3σ can bind to p53 and leads to p53 stabilization and enhances transcriptional activity of p53 (31). Therefore, as a target gene of p53, 14-3-3σ appears to have a positive feedback effect on p53 activity. In addition to 14-3-3σ, several 14-3-3 isoforms, including 14-3-3γ have been demonstrated to interact with p53 both in vitro and in vivo and in turn, this interaction increases the DNA binding activity of p53 (32,33).

In a previous study, we found that the 14-3-3γ protein is elevated in human lung cancerous tissues (16) and overexpression of this protein in lung cancer cells affects cell cycle progression and results in DNA polyploidization (34). In addition, wild-type p53 can bind the 14-3-3γ promoter and reduce its mRNA (35). In the present study we investigated the expression of p53 and 14-3-3γ in human lung cancer tissues and discovered that 14-3-3γ expression is significantly correlated with p53 mutation. Further investigation using cultured colon and lung cancer cells showed wild-type of p53 but not mutant p53 (R175H) could suppress 14-3-3γ expression. Importantly, inhibition of 26S proteasome by MG132 blocked the reduction of 14-3-3γ by p53. These results indicate that p53 negatively regulates 14-3-3γ by stimulating proteasome-mediated 14-3-3γ protein degradation.

Materials and methods

Antibodies and reagents

Anti-14-3-3γ (specific) antibody was obtained from Santa Cruz Biotechnology (Santa Cruz, CA, USA). Antibodies to flag the β-actin were purchased from Sigma (St. Louis, MO, USA). Anti-p53 (DO-1) and GFP antibodies were obtained from Oncogene (La Jolla, CA, USA) and BD Pharmingen (San Diego, CA, USA), respectively. Cycloheximide and MG132 were purchased from Calbiochem (La Jolla, CA, USA).

Plasmid construction and cell transfection

cDNA encoding human 14-3-3γ was cloned behind the CMV promoter in pCMV-Tag2 (with flag tag at N-terminal) expression vector (Stratagene, La Jolla, CA, USA). Human non-small cell lung carcinoma cell line H358 (p53 null) from the American Type Culture Collection (ATCC, Rockville, MD, USA) was cultured in Dulbecco’s modified Eagle’s medium (DMEM; Gibco-BRL) supplemented with 10% fetal bovine serum. Transfection was conducted using LipoTAXI mammalian transfection kit (Stratagene, La Jolla, CA, USA) according to the manufacturer’s instructions. In brief, 3×105 H358 cells were seeded in 60-mm dish and the cells cultured until they reached a confluence of 50–70%. The cells were washed with serum/antibiotic-free DMEM medium prior to transfection. Plasmid pCMV-flag-14-3-3γ (5 μg) purified with Perfectprep Plasmid midi (Eppendorf, Hamburg, Germany) and 50 μl of LipoTAXI reagent diluted in 900 μl of serum/antibiotic-free DMEM medium were mixed gently and incubated at room temperature for 30 min. After adding 1.5 ml of serum/antibiotic-free DMEM medium the mixture was placed onto the cells. Transfection was performed at 37°C in an incubator under 5% CO2 for 5 h and then further 2.5 ml DMEM containing 20% serum was added. After incubation overnight the DNA mixture was replaced by fresh, complete medium and the cells were incubated for 48 h and split into three 10-cm plates. When the cells grew to 95% confluence, 0.5 μg/ml of G418 (Invitrogen, Carlsbad, CA, USA) was added to medium to select the clones. After three weeks, 24 clones were collected and expression of 14-3-3γ protein was assessed by immunoblotting using anti-Flag antibody (Sigma).

Adenovirus construction and infections

Ad-GFP, Ad-p53 (wild-type) and Ad-p53 (mutant, R175H) expressed GFP, wild-type and mutant p53 were kindly provided by Dr Cyrus Vaziri (Boston University School of Medicine) and Dr Bernard Futscher (Arizona Cancer Center, The University of Arizona), respectively. HCT116 cells with p53 knocked out were plated in 60-mm culture dishes (in DMEM containing 10% fetal bovine serum) and infected by adding virus directly to the medium when the cultures had attained a confluence of ~60%. After 24-, 32- and 48-h infection, the cells were harvested and endogenous 14-3-3γ protein level was analyzed by immunoblotting. H358 cells stably expressing flag-14-3-3γ were also infected by adenovirus-GFP, wild-type and mutant p53 for 24 h and then treated with cycloheximide alone or together with proteasome inhibitor MG132. Eight and 16 h later, cells were lysed and exogenous flag-14-3-3γ was analyzed by immunoblotting.

The 14-3-3γ truncated constructs and protein expression

Human 14-3-3γ fragments (1:1-78 amino acid, N-terminal; 2:79-131 amino acid, central part; 3:132-184 amino acid, central part; and 4:185-247 amino acid, C-terminal) were amplified by PCR and inserted in-frame into pGEX2T vector (Amersham Pharmacia Biotech) for expression of GST fusion proteins. All constructs were sequenced to make sure the sequences were correct. The recombinant glutathione-S-transferase (GST) tagged proteins were expressed in E. coli strain DH5α with 0.5 mM isopropyl-β-L-thiogalactopyranoside (IPTG) at 37°C for 3 h. Cells were collected and lysed in B-PER bacterial protein extraction reagent (Pierce). After incubation on ice 30 min, the samples were centrifuged and the supernatants were purified using glutathione sepharose 4B beads (Amersham Pharmacia Biotech).

Glutathione-S-transferase pull-down assay and western blotting

Confluent cells (70%) were harvested and lysed in 0.5% Nonidet P-40 lysis buffer (50 mM Tris-Cl, pH 7.4, 0.25 M NaCl, 0.5% NP-40, 50 mM NaF and ptotease inhibitors). The pull-down assay was conducted by incubating cell extracts in lysis buffer with GST-14-3-3s or GST bound to 40 μl glutathione sepharose beads for 2 h at 4°C. After five washes with lysis buffer, the bound proteins were subjected to SDS-PAGE, transferred to nitrocellulose membrane, and immunoblotted with primary antibodies.

Protein analysis

Frozen human lung cancerous (squamous cell carcinoma and adenocarcinoma) and normal tissues were obtained from the Department of Pathogenic Biology, Affiliated Hospital of Jiangsu University (Zhenjiang, China). Frozen tissues and cultured cells were lysed in NP-40 lysis buffer containing 50 mM Tris-Cl (pH 7.4), 0.15 M NaCl, 0.5% NP-40, 1 mM DTT, 50 mM sodium fluoride, and 2 μl/ml protease inhibitor cocktail (Sigma). Protein concentrations were determined using the Bio-Rad protein assay kit (Bio-Rad Laboratories, Hercules, CA, USA) and 50 μg protein was resolved by electrophoresis on a 10% SDS-PAGE gel. The proteins were then transferred onto a nitrocellulose membrane and non-specific binds were blocked by incubating with 5% non-fat milk in TBST buffer (0.01 M Tris-Cl, pH 8.0, 0.15 M NaCl, 0.5% Tween-20) at room temperature for 1 h. The membrane was subjected to the indicated antibodies and the proteins were detected by the SuperSignal West Pico detection system (Pierce, Rockford, IL, USA).

Ubiquitination assay

H358 cells were co-transfected with Flag-tagged 14-3-3γ expression vector and His-tagged ubiquitin expression vector. At 16 h after transfection, cells were infected with Ad-GFP, Ad-p53 (wt) and Ad-p53 (mt) for 32 h and then harvested. Cells were lysed in NP-40 lysis buffer (50 mM Tris-Cl, pH 7.4, 0.25 M NaCl, 0.5% NP-40, 50 mM NaF and ptotease inhibitors). Co-immunoprecipitation was conducted using Anti-Flag M2 affinity gel (Sigma). Briefly, 200 μg of total protein in 500 μl lysis buffer was incubated with 20 μl Anti-Flag M2 affinity gel at 4°C overnight with rotation and the beads were then washed four times with lysis buffer. The proteins were eluted by boiling in SDS-PAGE sample buffer and applied to SDS-PAGE gels. 14-3-3γ ubiquitin was detected by immunoblotting using the anti-ubiquitin antibody (Upstate).

Statistical analysis

The Chi-squared test was performed to determine the correlation between mutant p53 and 14-3-3γ protein expression and P<0.05 is considered significant.

Results

14-3-3γ protein expression correlates with overexpression of p53 in human lung cancerous tissues

Our previous study indicates that high levels of 14-3-3γ protein exist in human lung cancer tissues as compared with the normal lung (16). Importantly, overexpression of 14-3-3γ protein can cause genomic instability by inducing polyploidization in human lung cancer cells (34). p53, a tumor suppressor protein, is reported to interact with several 14-3-3 isoforms including 14-3-3γ and this interaction appears to be functionally significant (32,33). In an attempt to understand the functional relationship between p53 and 14-3-3γ in lung cancers we determined the expression of p53 and 14-3-3γ by western blotting. In total 80 lung cancer tissues and 35 lung normal tissues were used in this experiment. A typical representative expression profile was shown in Fig. 1. As expected, we could not detect any signals of p53 in the 35 normal tissues. Consistent with our previous study, only one of 35 normal tissues had a band of 14-3-3γ. This normal tissue was probably contaminated with cancer cells. However, out of 80 cancer tissues there were 35 (43.8%) with p53 overexpression of (Fig. 1). In these 35 specimens, 33 expressed 14-3-3γ protein. Statistical analysis between 14-3-3γ and p53 proteins expression showed that 14-3-3γ expression significantly correlated with overexpression of p53 protein (P=0.0001; Table I). This result indicated that 14-3-3γ expression in lung cancers was dependent on overexpression of p53.

Table I

Expression of 14-3-3γ correlates with overexpression of p53 in human lung cancer tissues.

Table I

Expression of 14-3-3γ correlates with overexpression of p53 in human lung cancer tissues.

No. of cancer tissues% of 14-3-3γ expression in p53 overexpressed tissuesP-value
p53 positive35
14-3-3γ positive3394.3%0.0001
14-3-3γ negative2
Wild-type p53 protein suppresses the 4-3-3γ protein level

p53 acts as a transcription factor to activate or silence a number of downstream target genes (27,28). To investigate the effect of p53 on 14-3-3γ protein level we introduced both wild-type p53 and mutant p53 (R175H) into human colon cancer cell line HCT116 in which endogenous wild-type p53 gene had been knocked out. We infected the cells with adenovirus containing GFP, wild-type p53 (wt) and mutant p53 (mt) cDNA for 24, 32 and 48 h, and then the endogenous 14-3-3γ protein level was examined by immunoblot analysis using a specific antibody against 14-3-3γ. The same blot was also probed with antibodies against GFP and p53 to detect the efficiency of adenovirus infection. Anti-human β-actin antibody was also used on the same blot to control for variations in loading. The results showed that after 32 h the wild-type p53 caused a considerable decrease (~50%) in 14-3-3γ protein level (Fig. 2). However, neither GFP nor mutant p53 (R175H) had any effect on levels of 14-3-3γ in HCT116 cells (p53−/−). Hence, wild-type p53, but not mutant p53 (R175H) suppressed endogenous 14-3-3γ protein levels in colon cancer cells.

p53 suppresses 14-3-3γ through protein degradation by proteasome

Like other genes, 14-3-3γ regulation may occur at multiple levels, including transcription, post-transcriptional regulation of mRNA, translation and post-translational modification (36). Our previous study demonstrated that one of mechanism of p53 suppression of 14-3-3γ is through binding its promoter and inhibiting 14-3-3γ transcription (35). In order to clarify whether p53 suppresses 14-3-3γ post-transcription we stably transfected 14-3-3γ with Flag tag into human lung cancer cell line H358 which is p53 null and then examine whether p53 suppresses this exogenous 14-3-3γ protein. H358 cells which constitutively express Flag-14-3-3γ protein were infected by adenovirus-GFP, wild-type p53 and mutant p53 (R175H) for 24 h and cycloheximide was added to inhibit protein synthesis. At 0, 8 and 24 h after cycloheximide treatment, the Flag-14-3-3γ protein levels were determined by immunoblot analysis using an antibody against Flag. Similarly to the effect of p53 on the endogenous 14-3-3γ in colon cancer cell line HCT116, wild-type p53 decreased 50% of Flag-14-3-3γ protein (Fig. 3). This result clearly indicates that p53 can negatively regulate 14-3-3γ by post-translational modification.

p53 suppression of 14-3-3γ could be through stimulating proeasome-mediated protein degradation

To determine whether p53 mediated 14-3-3γ reduction by stimulating proteasomal degradation of 14-3-3γ a specific inhibitor of proteasomal activity, MG132, was used and its effect on p53-mediated 14-3-3γ reduction was examined. The results showed that MG132 treatment completely abolished the p53-mediated reduction of 14-3-3γ protein (Fig. 4) which suggests that p53-mediated suppression of 14-3-3γ is accomplished by stimulating the process of proteasomal degradation of the protein.

C-terminal of 14-3-3γ binds to p53

It has been reported that wild-type of p53 can bind to 14-3-3γ (32,33). This interaction may lead to proteasomal degradation of 14-3-3 protein. However, the detailed binding domain of 14-3-3γ is still unclear. To further explore the possible region of 14-3-3γ that is involved in binding to p53, we made GST-14-3-3γ truncated fusion proteins including N-terminal, GST-14-3-3γ1 (1-78 amino acids); central regions, GST-14-3-3γ2 and 3 (79-131 and 132-184 amino acids); and C-terminal GST-14-3-3γ4 (185-247 amino acids) (Fig. 5A). HCT116 cells were lysed and pull-down assay experiment was performed to determine the interaction. Fig. 5B showed that the C-terminal region of 14-3-3γ bound to p53 very efficiently compared with other domains, demonstrating that p53 could interact with C-terminal of 14-3-3γ.

p53 enhances 14-3-3γ ubiquitination

Since ubiquitination is required in the process of protein degradation by 26S protesome which can be blocked by MG132, we next determined whether p53 increases 14-3-3γ protein ubiquitin. H358 cells were co-transfected with Flag-tagged 14-3-3γ expression vector and His-tagged ubiquitin expression vector and then infected with Ad-GFP, Ad-p53 (wt) and Ad-p53 (mt) viruses. After immunoprecipitation by anti-Flag antibody, 14-3-3γ ubiquitination was evaluated, and Fig. 6 clearly shows that wild-type p53 stimulated 14-3-3γ ubiquitination in vivo. Taken together, these data indicated that p53 could bind to 14-3-3γ, and enhanced its ubiquitin and proteasome-mediated degradation.

Discussion

14-3-3γ protein is overexpressed in human lung cancer tissues (16) and plays a role in the development of cancer (37). Notably, 14-3-3γ also has been demonstrated to interact with tumor suppression protein p53 and enhance transcriptional activity of p53 (33). Therefore, it is important to characterize the expression regulation of 14-3-3γ. In the present study, we investigated the expression profile of both 14-3-3γ and p53 in human lung cancer tissues and found that 14-3-3γ protein expression correlated with overexpression of p53. Since wild-type p53 protein level is usually too low to detect we supposed the p53 see in the bands on the blot to be mutated. Therefore, it is possible that 14-3-3γ expression correlated with p53 mutation in lung cancer patients. Ecotopic expression experiments showed wild-type p53, but not mutant p53 (R175H) could reduce both endogenous and exogenous 14-3-3γ protein levels in colon and lung cancer cells. In a previous study we showed that wild-type p53 inhibited 14-3-3γ on mRNA level (35). In the present study, we further demonstrated that p53 also can suppress 14-3-3γ by stimulating proteasome-mediated 14-3-3γ protein degradation. Together, these data indicate that p53 negatively regulate 14-3-3γ on both of transcriptional and post-translational level.

p53 has been studied intensely for its function as a genome stability guardian by regulating cell cycle arrest, apoptosis and DNA repair after DNA damage (26,38). It is also well known that p53 is the most commonly mutated gene with up to 50% mutations in different types of cancers. Consistent with previous studies, we found here p53 was overexpressed in 35 out of 80 human lung cancerous tissues. Importantly, high expression of 14-3-3γ was observed in 94% of these tissues with overexpression of p53 (33 of 35). Because overexpression of 14-3-3γ protein can cause DNA polyploidization in human lung cancer (34) and induced cell oncogenic transformation (37), it is possible that p53 guards genome stability and prevents tumorigenesis through inhibiting 14-3-3γ protein in the lung. Therefore, 14-3-3γ could be a novel target of p53 for maintaining genomic stability.

As a tumor suppressor gene, 14-3-3σ expression levels are significantly reduced or totally lost in a number of cancers, such as oral squamous cell carcinomas (39), primary bladder tumors (40), most types of breast cancer (41), gastric cancer (42) and hepatocellular carcinoma (43). One of the mechanisms resulting in 14-3-3σ reduction in cancers is due to p53 mutation because 14-3-3σ is transactivated directly by p53 (29). On the contrary, p53 negatively regulates 14-3-3γ by promoting its protein degradation. Taken together, these data suggest that p53 has diverse effects on 14-3-3 proteins in an isoform specific manner. p53 has been demonstrated to be mutated in almost half of all human cancers. In response to this, tumor suppression protein 14-3-3σ will be lost and the potential oncoprotein 14-3-3γ will progress. Therefore, p53 may act as the center of a complex network in tumorigenesis by regulating differently the 14-3-3 family proteins.

Recently, 14-3-3 proteins were identified to bind to an E3 ubiquitin-protein ligase, tripartite motif-containing protein 32 (TRIM32) and to prevent TRIM32 auto-ubiquitylation (44). Notably, Sato et al (45) also discovered the ubiquitin ligase ATL31 associated with and ubiquitinated 14-3-3 proteins for degradation via the ubiquitin-proteasome system during the response to cellular carbon (C)/nitrogen (N) status in Arabidopsis. Therefore, it is possible that p53 can regulate the interaction between 14-3-3γ and ubiquitin ligase. However, the mechanism of 14-3-3γ ubiquitination, especially enhanced by p53 is still unclear and further studies are needed.

Acknowledgements

The present study is supported by the fund from the Health Administration of Jiangsu Province.

References

1 

Aitken A: 14-3-3 proteins: a historic overview. Semin Cancer Biol. 16:162–172. 2006. View Article : Google Scholar : PubMed/NCBI

2 

Dougherty MK and Morrison DK: Unlocking the code of 14-3-3. J Cell Sci. 117:1875–1884. 2004. View Article : Google Scholar : PubMed/NCBI

3 

Hermeking H: The 14-3-3 cancer connection. Nat Rev Cancer. 3:931–943. 2003. View Article : Google Scholar

4 

Hermeking H and Benzinger A: 14-3-3 proteins in cell cycle regulation. Semin Cancer Biol. 16:183–192. 2006. View Article : Google Scholar : PubMed/NCBI

5 

Zhang Y, Karas M, Zhao H, Yakar S and LeRoith D: 14-3-3-sigma mediation of cell cycle progression is p53-independent in response to insulin-like growth factor-I receptor activation. J Biol Chem. 279:34353–34360. 2004. View Article : Google Scholar : PubMed/NCBI

6 

Kasahara K, Goto H, Enomoto M, Tomono Y, Kiyono T and Inagaki M: 14-3-3gamma mediates Cdc25A proteolysis to block premature mitotic entry after DNA damage. EMBO J. 29:2802–2812. 2010. View Article : Google Scholar : PubMed/NCBI

7 

Du J, Chen L, Luo X, Shen Y, Dou Z, Shen J, Cheng L, Chen Y, Li C, Wang H and Yao X: 14-3-3zeta cooperates with phosphorylated Plk1 and is required for correct cytokinesis. Front Biosci (Schol Ed). 4:639–650. 2012. View Article : Google Scholar

8 

Masters SC and Fu H: 14-3-3 proteins mediate an essential antiapoptotic signal. J Biol Chem. 276:45193–45200. 2001. View Article : Google Scholar : PubMed/NCBI

9 

Mhawech P: 14-3-3 proteins - an update. Cell Res. 15:228–236. 2005. View Article : Google Scholar : PubMed/NCBI

10 

Yan Y, Xu Y, Gao YY, Zong ZH, Zhang Q, Li C and Wang HQ: Implication of 14-3-3epsilon and 14-3-3theta/tau in proteasome inhibition-induced apoptosis of glioma cells. Cancer Sci. 104:55–61. 2005. View Article : Google Scholar

11 

Han DC, Rodriguez LG and Guan JL: Identification of a novel interaction between integrin beta1 and 14-3-3beta. Oncogene. 20:346–357. 2001. View Article : Google Scholar : PubMed/NCBI

12 

Rodriguez LG and Guan JL: 14-3-3 regulation of cell spreading and migration requires a functional amphipathic groove. J Cell Physiol. 202:285–294. 2005. View Article : Google Scholar

13 

Luk SC, Ngai SM, Tsui SK, Fung KP, Lee CY and Waye MM: In vivo and in vitro association of 14-3-3 epsilon isoform with calmodulin: implication for signal transduction and cell proliferation. J Cell Biochem. 73:31–35. 1999. View Article : Google Scholar : PubMed/NCBI

14 

Matitau AE, Gabor TV, Gill RM and Scheid MP: MEKK2 association with 14-3-3 regulates activation of c-Jun N-terminal kinase. J Biol Chem. 288:28293–28302. 2013. View Article : Google Scholar : PubMed/NCBI

15 

Radhakrishnan VM, Putnam CW and Martinez JD: Activation of phosphatidylinositol 3-kinase (PI3K) and mitogen-activated protein kinase (MAPK) signaling and the consequent induction of transformation by overexpressed 14-3-3gamma protein require specific amino acids within 14-3-3gamma N-terminal variable region II. J Biol Chem. 287:43300–43311. 2012. View Article : Google Scholar : PubMed/NCBI

16 

Qi W, Liu X, Qiao D and Martinez JD: Isoform-specific expression of 14-3-3 proteins in human lung cancer tissues. Int J Cancer. 113:359–363. 2005. View Article : Google Scholar

17 

Fan T, Li R, Todd NW, Qiu Q, Fang HB, Wang H, Shen J, Zhao RY, Caraway NP, Katz RL, Stass SA and Jiang F: Up-regulation of 14-3-3zeta in lung cancer and its implication as prognostic and therapeutic target. Cancer Res. 67:7901–7906. 2007. View Article : Google Scholar : PubMed/NCBI

18 

Titus MA, Tan JA, Gregory CW, Ford OH, Subramanian RR, Fu H, Wilson EM, Mohler JL and French FS: 14-3-3η amplifies androgen receptor actions in prostate cancer. Clin Cancer Res. 15:7571–7581. 2009. View Article : Google Scholar : PubMed/NCBI

19 

Han B, Xie H, Chen Q and Zhang JT: Sensitizing hormone-refractory prostate cancer cells to drug treatment by targeting 14-3-3sigma. Mol Cancer Ther. 5:903–912. 2006. View Article : Google Scholar : PubMed/NCBI

20 

Li Z, Dong Z, Myer D, Yip-Schneider M, Liu J, Cui P, Schmidt CM and Zhang JT: Role of 14-3-3sigma in poor prognosis and in radiation and drug resistance of human pancreatic cancers. BMC Cancer. 10:5982010. View Article : Google Scholar

21 

Naidoo K, Jones R, Dmitrovic B, Wijesuriya N, Kocher H, Hart IR and Crnogorac-Jurcevic T: Proteome of formalin-fixed paraffin-embedded pancreatic ductal adenocarcinoma and lymph node metastases. J Pathol. 226:756–763. 2012. View Article : Google Scholar

22 

Nishimura Y, Komatsu S, Ichikawa D, Nagata H, Hirajima S, Takeshita H, Kawaguchi T, Arita T, Konishi H, Kashimoto K, Shiozaki A, Fujiwara H, Okamoto K, Tsuda H and Otsuji E: Overexpression of YWHAZ relates to tumor cell proliferation and malignant outcome of gastric carcinoma. Br J Cancer. 108:1324–1331. 2013. View Article : Google Scholar : PubMed/NCBI

23 

O’Dwyer D, Ralton LD, O’Shea A and Murray GI: The proteomics of colorectal cancer: identification of a protein signature associated with prognosis. PLoS One. 6:e277182011. View Article : Google Scholar

24 

Yang X, Cao W, Zhang L, Zhang W, Zhang X and Lin H: Targeting 14-3-3zeta in cancer therapy. Cancer Gene Ther. 19:153–159. 2012. View Article : Google Scholar

25 

Kaufmann WK and Paules RS: DNA damage and cell cycle checkpoints. FASEB J. 10:238–247. 1996.PubMed/NCBI

26 

Mendoza-Rodriguez CA and Cerbon MA: Tumor suppressor gene p53: mechanisms of action in cell proliferation and death. Rev Invest Clin. 53:266–273. 2001.(In Spanish).

27 

Yang X, Taylor L and Polgar P: p53 down-regulates human brady-kinin B1 receptor gene expression. J Cell Biochem. 82:38–45. 2001. View Article : Google Scholar : PubMed/NCBI

28 

el-Deiry WS: Regulation of p53 downstream genes. Semin Cancer Biol. 8:345–357. 1998. View Article : Google Scholar

29 

Hermeking H, Lengauer C, Polyak K, He TC, Zhang L, Thiagalingam S, Kinzler KW and Vogelstein B: 14-3-3 sigma is a p53-regulated inhibitor of G2/M progression. Mol Cell. 1:3–11. 1997. View Article : Google Scholar

30 

Laronga C, Yang HY, Neal C and Lee MH: Association of the cyclin-dependent kinases and 14-3-3 sigma negatively regulates cell cycle progression. J Biol Chem. 275:23106–23112. 2000. View Article : Google Scholar : PubMed/NCBI

31 

Yang HY, Wen YY, Chen CH, Lozano G and Lee MH: 14-3-3 sigma positively regulates p53 and suppresses tumor growth. Mol Cell Biol. 23:7096–7107. 2003. View Article : Google Scholar : PubMed/NCBI

32 

Rajagopalan S, Sade RS, Townsley FM and Fersht AR: Mechanistic differences in the transcriptional activation of p53 by 14-3-3 isoforms. Nucleic Acids Res. 38:893–906. 2010. View Article : Google Scholar :

33 

Stavridi ES, Chehab NH, Malikzay A and Halazonetis TD: Substitutions that compromise the ionizing radiation-induced association of p53 with 14-3-3 proteins also compromise the ability of p53 to induce cell cycle arrest. Cancer Res. 61:7030–7033. 2001.PubMed/NCBI

34 

Qi W, Liu X, Chen W, Li Q and Martinez JD: Overexpression of 14-3-3gamma causes polyploidization in H322 lung cancer cells. Mol Carcinog. 46:847–856. 2007. View Article : Google Scholar : PubMed/NCBI

35 

Radhakrishnan VM, Putnam CW, Qi W and Martinez JD: P53 suppresses expression of the 14-3-3 gamma oncogene. BMC Cancer. 11:3782011. View Article : Google Scholar : PubMed/NCBI

36 

Qiao D, Gaitonde SV, Qi W and Martinez JD: Deoxycholic acid suppresses p53 by stimulating proteasome-mediated p53 protein degradation. Carcinogenesis. 22:957–964. 2001. View Article : Google Scholar : PubMed/NCBI

37 

Radhakrishnan VM and Martinez JD: 14-3-3gamma induces oncogenic transformation by stimulating MAP kinase and PI3K signaling. PLoS One. 5:e114332010. View Article : Google Scholar : PubMed/NCBI

38 

Golubovskaya VM and Cance WG: Targeting the p53 pathway. Surg Oncol Clin N Am. 22:747–764. 2013. View Article : Google Scholar : PubMed/NCBI

39 

Bhawal UK, Sugiyama M, Nomura Y, Kuniyasu H and Tsukinoki K: Loss of 14-3-3 sigma protein expression and presence of human papillomavirus type 16 E6 in oral squamous cell carcinoma. Arch Otolaryngol Head Neck Surg. 134:1055–1059. 2008. View Article : Google Scholar : PubMed/NCBI

40 

Ostergaard M, Rasmussen HH, Nielsen HV, Vorum H, Orntoft TF, Wolf H and Celis JE: Proteome profiling of bladder squamous cell carcinomas: identification of markers that define their degree of differentiation. Cancer Res. 57:4111–4117. 1997.PubMed/NCBI

41 

Ferguson AT, Evron E, Umbricht CB, Pandita TK, Chan TA, Hermeking H, Marks JR, Lambers AR, Futreal PA, Stampfer MR and Sukumar S: High frequency of hypermethylation at the 14-3-3 sigma locus leads to gene silencing in breast cancer. Proc Natl Acad Sci USA. 97:6049–6054. 2000. View Article : Google Scholar : PubMed/NCBI

42 

Suzuki H, Itoh F, Toyota M, Kikuchi T, Kakiuchi H and Imai K: Inactivation of the 14-3-3 sigma gene is associated with 5′ CpG island hypermethylation in human cancers. Cancer Res. 60:4353–4357. 2000.PubMed/NCBI

43 

Iwata N, Yamamoto H, Sasaki S, Itoh F, Suzuki H, Kikuchi T, Kaneto H, Iku S, Ozeki I, Karino Y, Satoh T, Toyota J, Satoh M, Endo T and Imai K: Frequent hypermethylation of CpG islands and loss of expression of the 14-3-3 sigma gene in human hepatocellular carcinoma. Oncogene. 19:5298–5302. 2000. View Article : Google Scholar : PubMed/NCBI

44 

Ichimura T, Taoka M, Shoji I, Kato H, Sato T, Hatakeyama S, Isobe T and Hachiya N: 14-3-3 proteins sequester a pool of soluble TRIM32 ubiquitin ligase to repress autoubiquitylation and cytoplasmic body formation. J Cell Sci. 126:2014–2026. 2013. View Article : Google Scholar : PubMed/NCBI

45 

Sato T, Maekawa S, Yasuda S, Domeki Y, Sueyoshi K, Fujiwara M, Fukao Y, Goto DB and Yamaguchi J: Identification of 14-3-3 proteins as a target of ATL31 ubiquitin ligase, a regulator of the C/N response in Arabidopsis. Plant J. 68:137–146. 2011. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

February-2015
Volume 46 Issue 2

Print ISSN: 1019-6439
Online ISSN:1791-2423

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Chen D, Dai D, Hua Y and Qi W: p53 suppresses 14-3-3γ by stimulating proteasome-mediated 14-3-3γ protein degradation. Int J Oncol 46: 818-824, 2015
APA
Chen, D., Dai, D., Hua, Y., & Qi, W. (2015). p53 suppresses 14-3-3γ by stimulating proteasome-mediated 14-3-3γ protein degradation. International Journal of Oncology, 46, 818-824. https://doi.org/10.3892/ijo.2014.2740
MLA
Chen, D., Dai, D., Hua, Y., Qi, W."p53 suppresses 14-3-3γ by stimulating proteasome-mediated 14-3-3γ protein degradation". International Journal of Oncology 46.2 (2015): 818-824.
Chicago
Chen, D., Dai, D., Hua, Y., Qi, W."p53 suppresses 14-3-3γ by stimulating proteasome-mediated 14-3-3γ protein degradation". International Journal of Oncology 46, no. 2 (2015): 818-824. https://doi.org/10.3892/ijo.2014.2740