Purpose: 14-3-3σ is an intracellular, dimeric, phosphoserine binding protein that is expressed in epithelial cells and involved in cancer development. In this study, we examined the expression of 14-3-3σ and evaluated its clinical significance in colorectal carcinoma.

Experimental Design: Expression of 14-3-3σ was analyzed by Western blot in nine colorectal carcinoma cell lines, eight paired colorectal carcinoma tissues, and normal mucosas. Immunohistochemistry was used to evaluate expression of 14-3-3σ in tissues of 121 colorectal carcinoma patients and to correlate it with clinical parameters.

Results: Western blot analysis of colorectal carcinoma cell lines and tissues revealed strong 14-3-3σ expression in four of eight cell lines and 14-3-3σ overexpression in carcinomas compared with normal mucosa in six of eight colorectal carcinoma tissue pairs. Immunohistochemical analysis revealed 14-3-3σ overexpression in 38.8% of colorectal carcinoma samples. Furthermore, highly positive immunoreactivity was significantly correlated with tumor differentiation (P < 0.001) and pT stage (P < 0.003). In Kaplan-Meier analysis, 14-3-3σ overexpression was associated with a significantly decreased survival time compared with negatively stained or low stained cases (P < 0.0096). In multivariate regression analysis, 14-3-3σ expression emerged as a significant independent parameter (P < 0.037).

Conclusions: These results provide evidence that 14-3-3σ expression increases during carcinoma progression in a subset of colorectal carcinoma. The overexpression of this antigen identifies patients at high risk. It is tempting to suggest that 14-3-3σ overexpression either promotes tumor proliferation and/or prevents apoptotic signal transduction in colorectal carcinoma. Thus, targeting 14-3-3σ might be a new therapeutic strategy in colorectal carcinoma.

14-3-3σ (also called stratifin) belongs to a family of abundant, widely expressed 28 to 33 kDa acidic polypeptides that spontaneously self-assemble as homodimers or heterodimers. There are seven closely related genes, encoding β, ε, η, γ, τ, ζ, and σ isoforms, that are conserved across mammalian species. They can bind to >100 functionally diverse cellular proteins and thereby play an important role in various cellular processes like signal transduction, cell cycle regulation, apoptosis, cytoskeleton organization, and malignant transformation (for a recent review, see refs. 13). 14-3-3 proteins contain no catalytic domain or function and a number of studies showed that binding of 14-3-3 requires target protein phosphorylation. Each monomer within the dimeric 14-3-3 molecule binds substrates through a binding cleft that preferentially recognizes proteins containing the optimal phosphoserine/phosphothreonine sequence motifs RSx[pS/pT]xP and RxΦx[pS/pT]xP (where uppercase bold characters denote the most highly conserved positions, the prefix p denotes a phosphorylated amino acid, Φ denotes aromatic or aliphatic amino acids and x represents any amino acid; refs. 4, 5). In addition, 14-3-3 proteins regulate enzyme activity and may act as localization anchors, controlling the subcellular localization of proteins. Furthermore, 14-3-3 proteins can function as adaptors or scaffolds stimulating protein-protein interactions (6).

14-3-3σ was originally identified as a p53 inducible gene that is responsive to DNA-damaging agents (7). 14-3-3σ sequesters the mitotic initiation complex, cdc2-cyclin B1, from entering the nucleus, thus preventing initiation of mitosis (8). In this manner, 14-3-3σ induces G2 arrest and allows the repair of damaged DNA. Furthermore, 14-3-3σ can bind cyclin-dependent kinase-2 and cyclin-dependent kinase-4 (CDK2, CDK4) and thereby block the transition of the eukaryotic cell cycle (9). These findings define 14-3-3σ as a negative regulator of cell cycle progression.

14-3-3σ expression is lost in ∼95% of breast carcinomas by methylation-mediated scilencing (10). Additionally, it has been shown that hypermethylation of 14-3-3σ occurs at an early stage in the progression of invasive breast cancer (11). Hypermethylation of CpG islands and loss of 14-3-3σ expression was also frequently found in human hepatocellular carcinoma (12), oral carcinoma (13), lung cancer (14), and prostate cancer (15). In colorectal carcinoma, however, Suzuki et al. (16) found that the CpG island of 14-3-3σ was unmethylated. Very recently, it was shown that hypermethylation of 14-3-3σ promoter is a rare event in colorectal carcinoma (17). The colorectal carcinoma cell lines HCT116, Lovo, and Lim2405 were found to express 14-3-3σ RNA (7). However, proteomic analysis revealed down-regulation of 14-3-3σ in human colon adenoma (18). Given the importance of 14-3-3σ silencing in cancer progression, only few data are available on 14-3-3σ expression in colorectal carcinoma.

Here, we analyzed the prognostic significance of 14-3-3σ expression in colorectal carcinoma compared with common clinicopathologic parameters in 121 patients with a long follow-up period, including univariate and multivariate analysis.

Reagents and chemicals. Unless otherwise indicated, all reagents were purchased from Sigma (Vienna, Austria). Cell culture reagents (media, serum, and antibiotics) were obtained from PAA Laboratories (Linz, Austria). Murine anti–14-3-3σ monoclonal antibody (mAb) was obtained from Neomarkers (Fremont, CA). Mouse anti–α-tubulin mAb was obtained from Sigma and the sheep anti-mouse horseradish peroxidase (HRP)-IgG was purchased from Amersham Pharmacia (Freiburg, Germany).

Cell culture, cell proliferation. Experiments were conducted with established human colon cancer cell lines (HRT-18, Caco-2, Lovo, DLD-1, SW480, CX-1, HCT-15, and HT-29) obtained from DSMZ (Darmstadt, Germany) and cultivated as suggested by the supplier with 10% FCS and 1% penicillin/streptomycin. New cultures were reestablished from frozen stocks every 3 months. The HT-29F cell line, which is a subclone of HT29 resistant to high concentrations of 5-fluorouracil (5-FU; ref. 19), was obtained from Dr. H. Hendriks (Faculty of Veterinary Medicine, Department of Pathology, Utrecht University, Utrecht, the Netherlands). Determination of IC50 of 5-FU exponentially growing cells of cell line HT29, and HT-29F were seeded at 7 × 105 cells per flask in the presence of increasing concentrations of the drug. Cell growth was assessed after 3 days by cell counting in a CASY (Schärfe Systems, Reutlingen, Germany) and IC50 values were determined by nonlinear curve fitting of data according to the general dose-response curve.

Reverse transcription-PCR analysis. Genomic DNA free RNA was isolated with the SV total RNA isolation system (Promega, Madison, WI) according to the manufacturer's protocol. Reverse transcription-PCR (RT-PCR) was done as described earlier (20). The following 14-3-3σ–specific primers was used: forward 5′-AGAGCGAAACCTGCTCTCAG-3′ and reverse 5′-GGCATCTCCTTCTTGCTGAT-3′. For control, glyceraldehyde 3-phosphodehydrogenase cDNA was amplified. RT-PCR without reverse transcriptase was run in parallel to verify no genomic DNA contamination. All PCR products were visualized by electrophoresis and ethidium bromide staining in 2% agarose gels.

Immunoblot analysis. Cells were detached by trypsinization and washed in PBS. Lysates were prepared in ice-cold cell lysis buffer (21). Tissue samples were homogenized, centrifuged, and the supernatant was taken for immunoblots. Equivalent protein concentrations of 30 μg were resolved in 12% SDS polyacrylamide gels on a Minigel apparatus (Novex, Groningen, the Netherlands) and transferred to a nitrocellulose membrane (Schleicher&Schuell, Einbeck, Germany). Membranes were incubated with the primary antibodies: anti–14-3-3σ (1:200) and mouse anti–α-tubulin (1:2,000). After the washing steps, membranes were incubated with IgG-HRP sheep anti-mouse (1:1,000). Protein-antibody complexes were detected by enhanced chemiluminescence (Amersham Pharmacia).

Patients and tissues. Formalin-fixed and paraffin-embedded samples from colorectal cancer (n = 121) were obtained from Department of Pathology, University of Innsbruck, Austria, from 1994 to 2003. Clinical and pathologic data were documented prospectively and entered into a specific tumor registry after surgery and after follow-up (Table 1). Each case was classified according to pT stage and nodal status (pTNM system), histologic tumor type, tumor differentiation (grades 1-3), tumor localization (right colon, left colon, rectum), and tumor size (<2, 2-5, and >5 cm).

Table 1.

Expression of 14-3-3σ correlated with clinicopathologic parameters (n = 121)

ParameterAll cases (N)14-3-3σ Low positive/negative [n = 74 (61.2%)]14-3-3σ High positive [n = 47 (38.8%)]Correlating significance (P)*
Gender 121    
    Male 63 37 (58.7) 26 (41.3)  
    Female 58 37 (63.8) 21 (36.2) 0.568 
Age at surgery (y) 121    
    <60 39 21 (53.8) 18 (46.2)  
    ≥60 82 53 (64.6) 29 (35.4) 0.255 
Tumor size (cm) 109    
    <2 13 11 (84.6) 2 (15.4)  
    2-5 65 38 (58.5) 27 (41.5) 0.084 
    >5 31 15 (48.4) 16 (51.6)  
Tumor localization 117    
    Right colon 33 17 (51.5) 16 (48.5)  
    Left colon 52 33 (63.5) 19 (36.5) 0.335 
    Rectum 32 22 (68.8) 10 (31.3)  
Tumor differentiation 121    
    Grade 1 29 25 (86.2) 4 (13.8)  
    Grade 2 83 47 (56.6) 36 (43.4) 0.001 
    Grade 3 2 (22.2) 7 (77.8)  
pT stage 121    
    pT1 26 23 (88.5) 3 (11.5)  
    pT2 18 7 (38.9) 11 (61.1) 0.003 
    pT3 68 37 (54.4) 31 (45.6)  
    pT4 7 (77.8) 2 (22.2)  
Nodal status 121    
    pN0 69 44 (63.8) 25 (36.2)  
    pN1-4 52 30 (57.7) 22 (42.3) 0.497 
Liver metastases 121    
    No 75 50 (66.7) 25 (33.3)  
    Yes 46 24 (52.2) 22 (47.8) 0.112 
ParameterAll cases (N)14-3-3σ Low positive/negative [n = 74 (61.2%)]14-3-3σ High positive [n = 47 (38.8%)]Correlating significance (P)*
Gender 121    
    Male 63 37 (58.7) 26 (41.3)  
    Female 58 37 (63.8) 21 (36.2) 0.568 
Age at surgery (y) 121    
    <60 39 21 (53.8) 18 (46.2)  
    ≥60 82 53 (64.6) 29 (35.4) 0.255 
Tumor size (cm) 109    
    <2 13 11 (84.6) 2 (15.4)  
    2-5 65 38 (58.5) 27 (41.5) 0.084 
    >5 31 15 (48.4) 16 (51.6)  
Tumor localization 117    
    Right colon 33 17 (51.5) 16 (48.5)  
    Left colon 52 33 (63.5) 19 (36.5) 0.335 
    Rectum 32 22 (68.8) 10 (31.3)  
Tumor differentiation 121    
    Grade 1 29 25 (86.2) 4 (13.8)  
    Grade 2 83 47 (56.6) 36 (43.4) 0.001 
    Grade 3 2 (22.2) 7 (77.8)  
pT stage 121    
    pT1 26 23 (88.5) 3 (11.5)  
    pT2 18 7 (38.9) 11 (61.1) 0.003 
    pT3 68 37 (54.4) 31 (45.6)  
    pT4 7 (77.8) 2 (22.2)  
Nodal status 121    
    pN0 69 44 (63.8) 25 (36.2)  
    pN1-4 52 30 (57.7) 22 (42.3) 0.497 
Liver metastases 121    
    No 75 50 (66.7) 25 (33.3)  
    Yes 46 24 (52.2) 22 (47.8) 0.112 
*

Probability, P, from χ2 test.

Immunohistochemistry. 14-3-3σ expression was determined by immunohistochemistry using a murine monoclonal antibody (Neomarkers). Four-micrometer sections were cut from paraffin block, deparaffinized, rehydrated, and put into endogenous peroxidase-blocking solution. The sections were then boiled (86°C) for 15 minutes in 10 mmol/L citrate-buffer (pH 6.0) in a water bath. Thereafter, slides were incubated with the primary antibody (1:100) at 4°C overnight. After washing in TBS, the slides were incubated in biotinylated rabbit anti-mouse IgG (DAKO, Copenhagen, Denmark) at a dilution of 1:500, and detected with an ABC-peroxidase kit (Vector Laboratories, Bulingame, CA) and diaminobezidine as substrate. The tissue sections were washed in water, counterstained with Mayer's hematoxylin (Merck, Darmstadt, Germany), and coverslipped. Negative controls were run in parallel.

Evaluation of 14-3-3σ expression. Slides of cancer specimens were analyzed in parallel by two independent pathologists (P. Obrist and S. Stadlmann) who had no prior knowledge of the clinical data. A 14-3-3σ immunostaining score was calculated on the basis of a proportion score and an intensity score as described previously (22). The proportion score reflects the estimated percentage of positively stained tumor cells (score 1, 0-10%; score 2, 10-50%; score 3, 51-80%; score 4, >80%). The intensity score represents the estimated staining intensity (score 0, no staining; score 1, weak; score 2, moderate; score 3, strong). 14-3-3σ overexpression was defined for tumors that show both a proportion score ≥3 and an intensity score ≥2.

Statistical analysis. All statistical analyses were done using SPSS 11.0 for Windows (SPSS, Inc., Chicago, IL). To trace correlations between 14-3-3σ expression and several clinicopathologic parameters, data were cross-tabulated and Fisher exact test was done. The correlation of 14-3-3σ staining with patient survival was evaluated using life tables constructed from survival data with Kaplan-Meier plots. Comparison of the different groups were done with the log-rank test. The end point in the present study was overall survival ranging from date of surgery until date of death or, if no information was documented, until date of last follow up information (=censored). Multivariate survival analysis was carried out on samples where all clinical parameters were available (n = 107) using the Cox proportional hazard model to evaluate the independent power of each variable. Results were considered significant when P < 0.05.

Expression of 14-3-3σ in colorectal carcinoma cell lines and tissues. To evaluate the expression of 14-3-3σ in colorectal carcinoma RT-PCR and Western blots of nine human colorectal carcinoma cell lines and Western blots of eight matched pairs of carcinoma and normal mucosa were done. RT-PCR revealed expression of 14-3-3σ in all nine colorectal carcinoma cell lines (Fig. 1A). Western blots determined a strong 14-3-3σ expression in CX-1, DLD-1, Caco-2, HT-29F, and Lovo cells; however, weak expression in HRT-18 and SW480 cells and no expression in HCT-15 and HT-29 cells were observed (Fig. 1B). In six tissue samples, as shown in Fig. 1B, 14-3-3σ expression was higher in carcinoma than in matched normal mucosa. In two samples (samples 5 and 6), 14-3-3σ expression was nearly similar in mucosa and carcinoma tissue (Fig. 1C). These results show differential 14-3-3σ expression between colorectal carcinoma cell lines on protein level but not on mRNA level. Furthermore, a significantly higher 14-3-3σ expression in carcinomas compared with normal mucosa was shown.

Fig. 1.

RT-PCR (A) and Western blot analysis of 14-3-3σ expression in colorectal carcinoma cell lines (B), and colorectal carcinoma tissue (c) and matched normal nonmalignant mucosa (m; C). Expression of 14-3-3σ mRNA was found in all analyzed cell lines (A). In contrast, 14-3-3σ protein was strongly expressed in cell lines CX-1, Caco-2, HT-29F, DLD-1, and Lovo; to a weaker extent in cell lines HRT-18 and SW480; and not expressed in cell lines HCT-15 and HT-29. (A) and (B) are representative of at least three independent experiments. In colorectal carcinoma tissue, a stronger 14-3-3σ signal was detected in carcinoma tissue compared with normal mucosa in six of eight analyzed patients. In two tissue pairs, the expression of 14-3-3σ was nearly similar in carcinoma and normal mucosa.

Fig. 1.

RT-PCR (A) and Western blot analysis of 14-3-3σ expression in colorectal carcinoma cell lines (B), and colorectal carcinoma tissue (c) and matched normal nonmalignant mucosa (m; C). Expression of 14-3-3σ mRNA was found in all analyzed cell lines (A). In contrast, 14-3-3σ protein was strongly expressed in cell lines CX-1, Caco-2, HT-29F, DLD-1, and Lovo; to a weaker extent in cell lines HRT-18 and SW480; and not expressed in cell lines HCT-15 and HT-29. (A) and (B) are representative of at least three independent experiments. In colorectal carcinoma tissue, a stronger 14-3-3σ signal was detected in carcinoma tissue compared with normal mucosa in six of eight analyzed patients. In two tissue pairs, the expression of 14-3-3σ was nearly similar in carcinoma and normal mucosa.

Close modal

Expression of 14-3-3σ in colorectal carcinoma tissue. To distinguish which cell type expresses 14-3-3σ in tissue samples, we did immunohistochemical analysis of 121 colorectal carcinoma tissues. One hundred sixteen (95.9%) of 121 patients with colorectal cancer showed a positive staining reaction for 14-3-3σ, whereas 5 (4.1%) specimens were completely 14-3-3σ negative (Fig. 2A). The atypical gland cxells showed membranous and cytoplasmic staining, which is most pronounced at the luminal surface (Fig. 2C′ and D′). The staining intensity was weak in 40 (34.5%), moderate in 43 (35.5%), and strong in 33 patients (27.3%; Fig. 2B-D). The mean percentage of stained tumor cells was 38.5 ± 17.8% (range 0-90%). Thirty-one specimens (25.6%) showed an area ≥50% with 14-3-3σ–positive tumor cells. The distribution of the percentage score (1-4; see Materials and Methods) was 8.3%, 71.9%, 18.2%, and 1.7%, respectively. The combination of staining intensity and percentage of stained tumor cells revealed 14-3-3σ overexpression in 47 cases (38.8%; for definition, see Materials and Methods).

Fig. 2.

14-3-3σ expression in normal colon mucosa and colorectal carcinoma as shown by immunohistochemistry. The figure shows moderately differentiated adenocarcinoma of the colon characterized by invasive malignant glands surrounded by desmoplastic stroma. Membranous and cytoplasmic 14-3-3σ–positive staining was found in the atypical gland cells, which is most pronounced at the luminal surface. No (A), weak (B), moderate (C), and strong (D) 14-3-3σ expression (original magnification, ×10). C′ and D′, ×20 magnification of (C) and (D).

Fig. 2.

14-3-3σ expression in normal colon mucosa and colorectal carcinoma as shown by immunohistochemistry. The figure shows moderately differentiated adenocarcinoma of the colon characterized by invasive malignant glands surrounded by desmoplastic stroma. Membranous and cytoplasmic 14-3-3σ–positive staining was found in the atypical gland cells, which is most pronounced at the luminal surface. No (A), weak (B), moderate (C), and strong (D) 14-3-3σ expression (original magnification, ×10). C′ and D′, ×20 magnification of (C) and (D).

Close modal

Clinicopathologic parameters and expression of 14-3-3σ are illustrated in Table 1. Univariate analysis revealed a significant correlation of 14-3-3σ overexpression with tumor differentiation (P = 0.001) and tumor stage (P = 0.003). A borderline association was found with increasing tumor size (P = 0.084). Colorectal carcinomas with small tumor size (<2 cm) had a lower percentage of 14-3-3σ overexpression (15.4%) when compared with carcinomas >5 cm (51.6%). The percentage of 14-3-3σ overexpression in colorectal carcinoma increases with tumor grade: Overexpression was observed in 13.8% of specimens with grade 1, 43.3% with grade 2, and 77.8% with grade 3. There was no significant correlation between the level of 14-3-3σ expression and other clinical and pathologic features, including sex, age, tumor localization, nodal status, and liver metastasis (Table 1). Survival analysis according to the Kaplan-Meier method revealed a significantly lower overall survival for patients with 14-3-3σ–overexpressing carcinomas (P = 0.0096; Fig. 3). Within this group, the 5-year survival rate was 56.2% compared with 73.6% in 14-3-3σ negative or low positive tumors. The median overall survival was 65 months (14-3-3σ overexpression) and not reached (14-3-3σ low expression), respectively. By multivariate analysis, 14-3-3σ overexpression was an independent prognostic marker for overall survival (P = 0.037). Of all clinicopathologic features, the presence of liver metastases was the most independent parameter predicting prognosis (P = 0.006; Table 2).

Fig. 3.

Kaplan-Meier estimates of overall survival for 121 patients with colorectral carcinomas grouped according to their 14-3-3σ expression. a, 14-3-3σ–negative or low positive cases (74). b, 14-3-3σ high positive cases (47).

Fig. 3.

Kaplan-Meier estimates of overall survival for 121 patients with colorectral carcinomas grouped according to their 14-3-3σ expression. a, 14-3-3σ–negative or low positive cases (74). b, 14-3-3σ high positive cases (47).

Close modal
Table 2.

Multivariate analysis (n = 107)

ParameterP*RR (95% CI)
Liver metastasis 0.006 2.880 (1.345-6.167) 
Age at surgery 0.035 2.200 (1.055-4.588) 
14-3-3σ Expression 0.037 1.940 (1.040-3.618) 
Nodal status 0.312 1.481 (0.691-3.173) 
Grading 0.471 1.568 (0.461-5.328) 
pT stage 0.697 0.844 (0.358-1.985) 
ParameterP*RR (95% CI)
Liver metastasis 0.006 2.880 (1.345-6.167) 
Age at surgery 0.035 2.200 (1.055-4.588) 
14-3-3σ Expression 0.037 1.940 (1.040-3.618) 
Nodal status 0.312 1.481 (0.691-3.173) 
Grading 0.471 1.568 (0.461-5.328) 
pT stage 0.697 0.844 (0.358-1.985) 

Abbreviations: RR, relative risk; 95% CI, 95% confidence interval for relative risk.

*

Probability, P, from Cox regression model.

Cell proliferation. 5-FU is still one of the most widely used agents in the first line therapy of colorectal carcinoma; therefore, we examined the effects of 5-FU on the growth of two colorectal carcinoma cell lines, which express 14-3-3σ (HT-29F) or not (HT-29). HT-29F is a subclone of HT29 cell line resistant to high concentration of 5-FU. The mean calculated antiproliferative IC50 values for 5-FU were 10.2 ± 0.31 μmol/L for HT-29F and 2.20 ± 0.18 μmol/L for HT-29, respectively. The results showed a significantly lower IC50 in the 14-3-3σ–negative than in the 14-3-3σ–positive cell line.

Among the seven distinct 14-3-3 genes, 14-3-3σ has been directly implicated in the etiology of human cancer (6). Recent studies have shown that 14-3-3σ is selectively lost by epigenetic hypermethylation of the gene promotor in breast cancer. In fact, loss of 14-3-3σ by gene methylation is reported to be the most consistent molecular alteration thus far discovered in breast cancer (10, 11). In contrast, methylation of 14-3-3σ was never detected in breast epithelial tissue derived from cancer-free patients. A second mechanism of 14-3-3σ loss in breast cancer is its targeting for ubiquitin-mediated proteolysis by Efp, an estrogen-dependent E3 ubiquitin ligase (23). 14-3-3σ inactivation by epigenetic silencing was also detected in a number of other cancer types, such as small cell lung cancer (14), ovarian cancer (24), hepatocellular carcinoma (12), prostate cancer (15), and gastric cancer (16). Furthermore, silencing of 14-3-3σ by methylation was detected in vulval and oral squamous cell carcinoma (13, 25) and in basal cell carcinomas (26). These data suggest that 14-3-3σ acts as a tumor suppressor and that its inactivation contributes to tumor progression.

In contrast to these findings, our results clearly show that 14-3-3σ is overexpressed in a subset of colorectal cancers and predicts poor overall survival in this patient group. Furthermore, 14-3-3σ overexpression was mainly found in dedifferentiated carcinomas and in more advanced tumor stages, indicating that 14-3-3σ is involved in colorectal carcinoma progression. In human colorectal carcinoma cells, 14-3-3σ is a p53 regulated gene and can induce cell cycle arrest at G2-M phase by a mechanism that involves its binding to and sequestering CDK1-cyclin B1 complexes in the cytoplasm (7). Experimentally induced loss of both 14-3-3σ alleles resulted in failure to maintain the G2 checkpoint after DNA damage, causing mitotic catastrophe as the cell entered mitosis (8). Our data suggest that 14-3-3σ expression is not solely dependent on intact p53 activity because most of the cell lines that express 14-3-3σ are p53 mutated (DLD-1, CX-1, Caco-2, SW480), whereas other cell lines are 14-3-3σ negative and also p53 mutated (HCT-15, HT-29, HT-29F). The p53 status of the Lovo cell line was wild type, whereas the status of HRT-18 (identical to HCT-8) cells is controversially reported (p53 status of different cell lines was obtained from recent literature; refs. 27, 28).

According to present and previous studies, epigenetic silencing of 14-3-3σ seems to be not involved in colorectal cancer because studies from Suzuki et al. (16) and Ide et al. (17) showed no CpG methylation of the 14-3-3σ promoter in colorectal carcinoma cell lines and carcinomas. Using RT-PCR, we found 14-3-3σ mRNA expression in all nine colorectal carcinoma cell lines. Similar results were also found in previous studies (7, 17). However, 14-3-3σ protein was differentially expressed in the analyzed colorectal carcinoma cell lines, suggesting the involvement of posttranscriptional regulatory mechanisms. Modifications of translational efficiency are increasingly reported in cancer and are controlled through a complex network of RNA/protein interactions involving recognition of mRNAs by RNA-binding proteins (29). It is tempting to suggest that a different composition of RNA/protein complexes in colorectal carcinoma cell lines might control translational efficiency of 14-3-3σ mRNA and account for the observed differences of 14-3-3σ expression in colorectal carcinoma cell lines. However, it is not known whether 14-3-3σ expression in colorectal carcinoma results from increased translational efficiency.

The specific inhibition of 14-3-3σ expression in various cancer types, but not in colorectal carcinoma, suggests a different role of 14-3-3σ in the transformation process in colorectal carcinoma than in other cancers. Cancer most commonly arises via two major abnormalities, which are uncontrolled cellular proliferation and lack of adequate cell death. 14-3-3σ overexpression might be involved in establishing of both abnormalities. Very recently, Zhang et al. showed that insulin-like growth factor-I increased 14-3-3σ expression and cell cycle progression in MCF7 breast cancer cells (30). These effects of insulin-like growth factor-I were p53 independent. Using small interfering RNA to reduce 14-3-3σ mRNA levels resulted in decrease of insulin-like growth factor-I–induced cell proliferation, demonstrating, at least in MCF7 cells, a positive link between 14-3-3σ expression and growth factor–induced cell cycle progression (30). In colorectal carcinoma, Ide et al. (17) provided evidence for a higher proliferative activity at the invasive front in 14-3-3σ–positive carcinomas than in 14-3-3σ–negative cases. These data suggest a possible growth advantage of 14-3-3σ–expressing cancer cells at least under specific growth conditions.

However, 14-3-3σ might also be involved in cell death regulation because 14-3-3σ was shown to prevent apoptosis in colorectal carcinoma cell lines through sequestration of BAX, a member of the bcl-2 family of apoptosis-regulating proteins, to the cytosol (31). Unbound BAX associates with mitochondria, mediates mitochondrial permeability transition, and subsequently results in cytochrome c release and apoptosis. Lack of 14-3-3σ in colorectal cancer cells sensitizes them to chemotherapy-induced apoptosis (31). By analysis of cell growth, we showed a positive correlation between 14-3-3σ expression and 5-FU sensitivity of colorectal carcinoma cells. Studies have correlated BAX and thymidylate synthase expression to 5-FU resistance of cancer cells (3234). By Western blot analysis, both proteins were equally expressed in HT-29 and HT-29F cell lines, suggesting that these proteins are not involved in the different sensitivity to 5-FU (data not shown). However, other proteins might be also involved in chemoresistance and, therefore, the suggested 14-3-3σ–mediated resistance to 5-FU chemotherapy remains to be proven.

Other functions of 14-3-3σ cannot be excluded because the different 14-3-3 monomers bind tightly to each other to form homodimers and mixed heterodimers and, therefore, display significant functional redundancy in vivo and in vitro (2). Except 14-3-3η and 14-3-3γ, all other members of the 14-3-3 protein family are expressed in all nine colorectal carcinoma cell lines (data not shown). Therefore, 14-3-3σ monomers may interact with other monomers of the 14-3-3 protein family to form functional heterodimers and activate tumor-promoting pathways. Finally, our results show overexpression of 14-3-3σ in ∼40% of colorectal cancer patients and predicts poor prognosis for these patients. Therefore, 14-3-3σ might be a potentially new target in colorectal carcinoma therapy.

Grant support: Österreichischen Krebshilfe-Krebsgesellschaft/Tirol.

The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

Note: A. Perathoner and D. Pirkebner contributed equally to this work.

We thank Martin Heitz for excellent technical assistance.

1
Dougherty MK, Morrison DK. Unlocking the code of 14-3-3.
J Cell Sci
2004
;
117
:
1875
–84.
2
Fu H, Subramanian RR, Masters SC. 14-3-3 proteins: structure, function, and regulation.
Annu Rev Pharmacol Toxicol
2000
;
40
:
617
–47.
3
Mackintosh C. Dynamic interactions between 14-3-3 proteins and phosphoproteins regulate diverse cellular processes.
Biochem J
2004
;
381
:
329
–42.
4
Muslin AJ, Xing H. 14-3-3 proteins: regulation of subcellular localization by molecular interference.
Cell Signal
2000
;
12
:
703
–9.
5
Yaffe MB, Rittinger K, Volinia S, et al. The structural basis for 14-3-3:phosphopeptide binding specificity.
Cell
1997
;
91
:
961
–71.
6
Hermeking H. The 14-3-3 cancer connection.
Nat Rev Cancer
2003
;
3
:
931
–43.
7
Hermeking H, Lengauer C, Polyak K, et al. 14-3-3σ is a p53-regulated inhibitor of G2/M progression.
Mol Cell
1997
;
1
:
3
–11.
8
Chan TA, Hermeking H, Lengauer C, Kinzler KW, Vogelstein B. 14-3-3σ is required to prevent mitotic catastrophe after DNA damage.
Nature
1999
;
401
:
616
–20.
9
Laronga C, Yang HY, Neal C, Lee MH. Association of the cyclin-dependent kinases and 14-3-3σ negatively regulates cell cycle progression.
J Biol Chem
2000
;
275
:
23106
–12.
10
Ferguson AT, Evron E, Umbricht CB, et al. High frequency of hypermethylation at the 14-3-3σ locus leads to gene silencing in breast cancer.
Proc Natl Acad Sci U S A
2000
;
97
:
6049
–54.
11
Umbricht CB, Evron E, Gabrielson E, Ferguson A, Marks J, Sukumar S. Hypermethylation of 14-3-3σ (stratifin) is an early event in breast cancer.
Oncogene
2001
;
20
:
3348
–53.
12
Iwata N, Yamamoto H, Sasaki S, et al. Frequent hypermethylation of CpG islands and loss of expression of the 14-3-3σ gene in human hepatocellular carcinoma.
Oncogene
2000
;
19
:
5298
–302.
13
Gasco M, Bell AK, Heath V, et al. Epigenetic inactivation of 14-3-3σ in oral carcinoma: association with p16(INK4a) silencing and human papillomavirus negativity.
Cancer Res
2002
;
62
:
2072
–6.
14
Osada H, Tatematsu Y, Yatabe Y, et al. Frequent and histological type-specific inactivation of 14-3-3σ in human lung cancers.
Oncogene
2002
;
21
:
2418
–24.
15
Cheng L, Pan CX, Zhang JT, et al. Loss of 14-3-3σ in prostate cancer and its precursors.
Clin Cancer Res
2004
;
10
:
3064
–8.
16
Suzuki H, Itoh F, Toyota M, Kikuchi T, Kakiuchi H, Imai K. Inactivation of the 14-3-3σ gene is associated with 5′ CpG island hypermethylation in human cancers.
Cancer Res
2000
;
60
:
4353
–7.
17
Ide M, Nakajima T, Asao T, Kuwano H. Inactivation of 14-3-3σ by hypermethylation is a rare event in colorectal cancers and its expression may correlate with cell cycle maintenance at the invasion front.
Cancer Lett
2004
;
207
:
241
–9.
18
Melis R, White R. Characterization of colonic polyps by two-dimensional gel electrophoresis.
Electrophoresis
1999
;
20
:
1055
–64.
19
Lesuffleur T, Kornowski A, Luccioni C, et al. Adaptation to 5-fluorouracil of the heterogeneous human colon tumor cell line HT-29 results in the selection of cells committed to differentiation.
Int J Cancer
1991
;
49
:
721
–30.
20
Pirkebner D, Fuetsch M, Wittmann W, et al. Reduction of intracellular pH inhibits constitutive expression of cyclooxygenase-2 in human colon cancer cells.
J Cell Physiol
2004
;
198
:
295
–301.
21
Gekle M, Schwerdt G, Freudinger R, et al. Ochratoxin A induces JNK activation and apoptosis in MDCK-C7 cells at nanomolar concentrations.
J Pharmacol Exp Ther
2000
;
293
:
837
–44.
22
Gastl G, Spizzo G, Obrist P, Dunser M, Mikuz G. Ep-CAM overexpression in breast cancer as a predictor of survival.
Lancet
2000
;
356
:
1981
–2.
23
Urano T, Saito T, Tsukui T, et al. Efp targets 14-3-3σ for proteolysis and promotes breast tumour growth.
Nature
2002
;
417
:
871
–5.
24
Kaneuchi M, Sasaki M, Tanaka Y, et al. Expression and methylation status of 14-3-3σ gene can characterize the different histological features of ovarian cancer.
Biochem Biophys Res Commun
2004
;
316
:
1156
–62.
25
Gasco M, Sullivan A, Repellin C, et al. Coincident inactivation of 14-3-3σ and p16INK4a is an early event in vulval squamous neoplasia.
Oncogene
2002
;
21
:
1876
–81.
26
Lodygin D, Yazdi AS, Sander CA, Herzinger T, Hermeking H. Analysis of 14-3-3σ expression in hyperproliferative skin diseases reveals selective loss associated with CpG-methylation in basal cell carcinoma.
Oncogene
2003
;
22
:
5519
–24.
27
Din FV, Dunlop MG, Stark LA. Evidence for colorectal cancer cell specificity of aspirin effects on NF κB signalling and apoptosis.
Br J Cancer
2004
;
91
:
381
–8.
28
Mariadason JM, Arango D, Shi Q, et al. Gene expression profiling-based prediction of response of colon carcinoma cells to 5-fluorouracil and camptothecin.
Cancer Res
2003
;
63
:
8791
–812.
29
Audic Y, Hartley RS. Post-transcriptional regulation in cancer.
Biol Cell
2004
;
96
:
479
–98.
30
Zhang Y, Karas M, Zhao H, Yakar S, LeRoith D. 14-3-3σ mediation of cell cycle progression is p53-independent in response to insulin-like growth factor-I receptor activation.
J Biol Chem
2004
;
279
:
34353
–60.
31
Samuel T, Weber HO, Rauch P, et al. The G2/M regulator 14-3-3σ prevents apoptosis through sequestration of Bax.
J Biol Chem
2001
;
276
:
45201
–6.
32
Nita ME, Nagawa H, Tominaga O, et al. 5-Fluorouracil induces apoptosis in human colon cancer cell lines with modulation of Bcl-2 family proteins.
Br J Cancer
1998
;
78
:
986
–92.
33
Zhang L, Yu J, Park BH, Kinzler KW, Vogelstein B. Role of BAX in the apoptotic response to anticancer agents.
Science
2000
;
290
:
989
–92.
34
Edler D, Glimelius B, Hallstrom M, et al. Thymidylate synthase expression in colorectal cancer: a prognostic and predictive marker of benefit from adjuvant fluorouracil-based chemotherapy.
J Clin Oncol
2002
;
20
:
1721
–8.