Purpose: Head and neck squamous cell carcinoma (HNSCC) represents the sixth most frequent type of cancer worldwide. However, the molecular genetic alterations underlying its malignant behavior and progression are little known. We showed previously that c-MYC directly activates the expression of the DNA double-strand break repair gene NBS1, and NBS1 overexpression contributes to transformation. Here, we investigate the role of NBS1 overexpression in HNSCC.

Experimental Design: Immunohistochemistry analysis of NBS1 expression was done in 81 locally advanced HNSCC patients. Real-time PCR and Western blot analysis were used to confirm immunohistochemistry results. Human hypopharyngeal cancer cell lines (FADU) with overexpressing NBS1 (FADUNBS) or inducible short interference RNA to repress endogenous NBS1 (FADUNBSi) were generated by stable transfection. Soft agar clonogenicity assay was used to determine the transformation activity. Western blot analysis and phosphatidylinositol 3-kinase (PI3K) assay were done to evaluate the signaling pathways that were involved.

Results: NBS1 overexpression was identified in 45% of advanced HNSCC patients. It was an independent marker of poor prognosis. NBS1 expression levels correlated with the transformation activity of FADU clones and also correlated with the phosphorylation levels of Akt and its downstream target mammalian target of rapamycin (mTOR). PI3K activity was increased in NBS1-overexpressing FADU clones. NBS1 overexpression also correlated with increased Akt phosphorylation levels in tumor samples.

Conclusions: Increased NBS1 expression is a significant prognostic marker of advanced HNSCC, and the underlying mechanism may involve the activation of the PI3K/Akt pathway.

Carcinomas of the head and neck, including cancers originating from the oral cavity, oropharynx, hypopharynx, and larynx, represent the sixth most frequent type of cancer in the world (1). They are ranked the fifth most lethal cause for male cancer patients in Taiwan, responsible for ∼1,560 deaths per year (2). More than 90% of head and neck cancers are squamous cell carcinoma (HNSCC; ref. 1), which are etiologically associated with betel nut (3), tobacco, and alcohol exposure (4). The treatment of early HNSCC includes surgical resection or radiotherapy. The organ preservation approach (induction chemotherapy followed by concurrent chemoradiotherapy) has recently been adopted to treat advanced HNSCC (5, 6). In spite of the improvement in diagnosis and management of HNSCC, long-term survival rates have improved only marginally over the past decade (1). The most commonly used prognostic markers of HNSCC are components of the staging system, including tumor stage and lymph node status (710). However, the accuracy of these clinical prognosticators is not as precise as desired (11). Molecular markers, such as ras, c-myc, epidermal growth factor receptor (EGFR), bcl-2, and p53, have been reported to be associated with clinical status and patient prognosis (1216). However, low incidence (such as c-myc and ras) and conflicting results from different reports (such as c-myc, bcl-2, p53, and EGFR) limit their application in the clinical setting (1, 1216). Developing new prognostic indicators to facilitate patient management is of the utmost importance.

Nijmegen breakage syndrome (NBS) is a chromosomal instability syndrome associated with cancer predisposition, radiosensitivity, and growth retardation (1719). The NBS gene product, NBS1, is a part of the hMre11 complex, which plays a central role in the DNA double-strand break repair (18, 19) and prevents double-strand break accumulation during chromosome replication (20). NBS1 carries out its checkpoint functions when it is phosphorylated by the ataxia telangiectasia mutated protein (ATM) following ionizing radiation (2123). However, rare or no mutations of NBS1 have been identified in certain types of human cancers (2427). The possible proliferation-inducing function of NBS1 is supported by the phenotypes of diminished expansion of the inner cell mass of mutant blastocysts (Nbs1 null) and cellular proliferation defects in Nbs1m/m mouse embryonic fibroblasts (2830). In addition, we showed previously that c-MYC, a dominant oncoprotein, directly activates NBS1 expression and that overexpression of NBS1 increases cell proliferation (31). Our recent results show that overexpression of NBS1 contributes to transformation through the activation of phosphatidylinositol 3-kinase (PI3K)/Akt (32). All these results implicated a role of NBS1 in the process of cell proliferation and tumorigenesis.

In this report, we showed that increased NBS1 expression is observed in a significant percentage (∼45%) of locally advanced HNSCC patients using immunohistochemistry, real-time PCR, and Western blot analysis. Increased NBS1 expression is a marker of aggressive HNSCC. Overexpression of NBS1 increases transformation in a head and neck cancer cell line (FADU). Increased NBS1 expression is correlated with the activation of PI3K/Akt pathway in cell lines and patient samples. These results show the oncogenic property of NBS1 overexpression and its role in the tumorigenesis and prognostic value of HNSCC.

Patients and treatment. From January 2000 to March 2003, 81 patients diagnosed as locally advanced squamous cell carcinoma of the oral cavity, oropharynx, hypopharynx, and larynx (T3-4NxM0) in Taipei Veterans General Hospital (Taipei, Taiwan) were enrolled in this study. Informed consent was obtained in writing before patient enrollment. This study has been approved by the Institutional Review Board of Taipei Veterans General Hospital. All patients were considered poor candidates for resection because surgery will cause significant functional impairment. Before the initiation of treatment, all patients received endoscopic biopsy for pathologic diagnosis. Study samples, including tumor and normal tissues, were obtained during diagnostic biopsy, and normal tissues were derived from neighboring site outside of the tumor. Both tumor and normal tissues for subsequent studies were confirmed by pathologists. The organ preservation protocol with two courses of induction chemotherapy (cisplatin + 5-fluorouracil) followed by concurrent chemoradiotherapy (including cisplatin + 5-fluorouracil and radiotherapy for primary tumor and neck up to 70 Gy in 35 fractions over 7 weeks) was applied to all patients. Evaluation of treatment response was done after induction chemotherapy and 2 months after chemoradiotherapy. The response criteria were based on RECIST criteria as described previously (33). Patients then received follow-up examinations every 3 months over the first year, every 4 months over the second year, and every 6 months thereafter. The medium follow-up period was 27 months (range, 18-56 months). The clinical characteristics of 81 HNSCC patients are illustrated in Table 1.

Table 1.

Characteristics of 81 advanced HNSCC patients

VariablesPatient, n (%)
Age  
    Range 38-74 
    Median 48 
Gender  
    Male 77 (95.1) 
    Female 4 (4.9) 
Site  
    Hypopharynx 28 (34.6) 
    Larynx 19 (23.5) 
    Oropharynx 11 (13.6) 
    Oral cavity 23 (28.4) 
T stage  
    T3 46 (56.8) 
    T4 35 (43.2) 
N stage  
    N0 31 (38.3) 
    N1-3 50 (61.7) 
Response to chemotherapy  
    Complete remission 31 (38.1) 
    Partial response 20 (24.7) 
    Stable disease 6 (7.5) 
    Progressive disease 24 (29.7) 
    Organ preservation rate 53 (65.4) 
VariablesPatient, n (%)
Age  
    Range 38-74 
    Median 48 
Gender  
    Male 77 (95.1) 
    Female 4 (4.9) 
Site  
    Hypopharynx 28 (34.6) 
    Larynx 19 (23.5) 
    Oropharynx 11 (13.6) 
    Oral cavity 23 (28.4) 
T stage  
    T3 46 (56.8) 
    T4 35 (43.2) 
N stage  
    N0 31 (38.3) 
    N1-3 50 (61.7) 
Response to chemotherapy  
    Complete remission 31 (38.1) 
    Partial response 20 (24.7) 
    Stable disease 6 (7.5) 
    Progressive disease 24 (29.7) 
    Organ preservation rate 53 (65.4) 

Immunohistochemistry. Sections (6 μm thick) of tumor and neighboring normal tissue were cut from the frozen specimens for immunohistochemistry analysis. The samples were fixed in acetone, air-dried, and subsequently bathed in TBS solution (pH 7.6). The endogenous peroxidase activity was blocked with 3% hydrogen peroxide. For NBS1 immunohistochemistry staining, a goat polyclonal anti-NBS1 antibody (Santa Cruz Biotechnology, Inc., Santa Cruz, CA) was used at the dilution of 1:100 and incubated for 1 hour. The immunohistochemistry of phosphorylated Akt was done with an immunohistochemistry-specific anti-phosphorylated Akt (Ser473) antibody (Cell Signaling Technology, Inc., Beverly, MA) at the dilution of 1:50 and incubated at 4°C overnight. After reacting with a biotinylated secondary antibody for 30 minutes, antigen-antibody reactions were visualized using streptavidin-horseradish peroxidase conjugate (DAKO LSAB kit; DAKO, Los Angles, CA), with 3-amino-9-ethylcarbazole as the chromogen. All slides were counterstained with hematoxylin.

Immunohistochemical scoring. All immunohistochemistry staining was independently scored by two experienced pathologists. If there was discordance with immunohistochemistry scoring, a pathologic peer review will be done to consolidate the result into a final score. The pathologists scoring the immunohistochemistry were blinded to the clinical data. The interpretation was done in five high-power views for each slide, and 100 cells per view were counted for analysis. Because normal tissues may have moderate NBS1 nucleus staining (34), the NBS1 immunohistochemistry scoring was defined on an scale ranging from 0 to +++ as illustrated (Fig. 1): 0, no appreciable staining in cells; +, only nucleus staining and no detectable cytoplasmic staining; ++, appreciable nucleus staining with cytoplasmic staining in <25% of cells; +++, significant nucleus staining and strong cytoplasmic staining (>25% of cells with cytoplasmic staining, which obscures cytoplasm and/or nucleus). Only NBS1 level +++ staining was defined as increased NBS1 expression.

Fig. 1.

Immunohistochemical analysis of NBS1 protein expression and localization in the normal upper aerodigestive epithelium (A and C) and tumor tissues (B and D-F) of HNSCC patients. Magnification, ×400. There is no NBS1 protein expression (immunohistochemistry level 0; A) or the presence of nucleus staining only (immunohistochemistry level +; C) in normal epithelium. Negative NBS1 staining (immunohistochemistry level 0; B), nucleus staining only (immunohistochemistry level +; D), or nucleus + weak cytoplasmic staining (immunohistochemistry level ++; E) was categorized as NBS1 nonoverexpression. Nucleus + strong cytoplasmic staining (immunohistochemistry level +++; F) was categorized as increased NBS1 expression. Black arrows, nucleus staining; yellow arrows, cytoplasmic staining.

Fig. 1.

Immunohistochemical analysis of NBS1 protein expression and localization in the normal upper aerodigestive epithelium (A and C) and tumor tissues (B and D-F) of HNSCC patients. Magnification, ×400. There is no NBS1 protein expression (immunohistochemistry level 0; A) or the presence of nucleus staining only (immunohistochemistry level +; C) in normal epithelium. Negative NBS1 staining (immunohistochemistry level 0; B), nucleus staining only (immunohistochemistry level +; D), or nucleus + weak cytoplasmic staining (immunohistochemistry level ++; E) was categorized as NBS1 nonoverexpression. Nucleus + strong cytoplasmic staining (immunohistochemistry level +++; F) was categorized as increased NBS1 expression. Black arrows, nucleus staining; yellow arrows, cytoplasmic staining.

Close modal

The scoring of phosphorylated Akt was as follows: 0, no appreciable staining in tumor cells; +, barely detectable staining in cytoplasm and/or nucleus compared with stromal elements; ++, readily appreciable brown staining distinctly marking tumor cell cytoplasm and/or nucleus; +++, dark brown staining in tumor cells completely obscuring cytoplasm and/or nucleus. The levels 0 and + were grouped as nonexpression, and levels ++ and +++ were grouped as overexpression according to the published report (35).

RNA purification from patient samples and real-time PCR analysis. We prepared RNA from fresh-frozen tumor and normal tissues using a RNeasy kit (Qiagen, Germany). Real-time PCR analysis was done using 1 μg total RNA isolated from tumor and neighboring normal tissues. Quantitative real-time PCR was done in a PRISM ABI7700 Sequence Detection System (Applied Biosystems, Foster City, CA) with the preset PCR program, and TBP (TATA box binding protein) was selected as an internal control (32). The primer sequences used in real-time PCR were NBS1 5′-ATGGAGGCCATATTTCCATGAC-3′ and 5′-CAAGCAGCCAGAACTTGGAAG-3′ and TBP 5′-TGGTGGTGTTGTGAGAAGATGG-3′ and 5′-CGGTGGGCACTTACAGAAGG-3′. cDNA (0.1 μg) and primers (80 μmol/L) were used for reaction in 1× SYBR Green Mixture (ABI) in a total volume of 50 μL. Increased NBS1 expression in tumor tissue was defined as >100% increase above normal tissue.

Protein extraction and Western blot analysis. For extraction of proteins from tumor tissues, 500 mg tumor samples were ground with rotor in 500 μL cell lysis buffer [50 mmol/L Tris (pH 7.5), 30 mmol/L MgSO4, 8 mmol/L EDTA, 2 mmol/L DTT, and 2% Triton X-100] containing protease inhibitors. Cell lysates were clarified by centrifugation at 13,000 rpm, 4°C for 10 minutes. The same lysis buffer was used to extract proteins from cultured cells. The protein content was determined by Bradford method (Bio-Rad Laboratories, Hercules, CA).

For Western blot analysis, 50 μg protein extracts from FADUCMV, FADUNBS, FADUNBSi, and FADUNBSim clones and tumor samples were loaded to 10% SDS-PAGE gels and transferred to nitrocellulose filters. The filters were probed with an anti-NBS1 antibody (NB100-143, Novus Biologicals, Inc., Littleton, CO), phosphorylated Akt (Ser473) antibody (Cell Signaling Technology), Akt antibody (Cell Signaling Technology), phosphorylated mammalian target of rapamycin (mTOR; Ser2448) antibody (Cell Signaling Technology), and an anti-β-actin antibody (Santa Cruz Biotechnology) or anti-γ-tubulin antibody (Santa Cruz Biotechnology) as loading control. Signals were developed using an enhanced chemiluminescence kit (Amersham Biosciences, Little Chalfont, United Kingdom). Data are representative of two or more experiments from independent cell cultures.

Plasmids, transfection, and cell lines. The human hypopharyngeal cancer cell lines FADU was obtained from American Type Culture Collection (Manassas, VA). FADU cells were cultured in RPMI 1640 with 10% heat-inactivated fetal bovine serum (FBS). The pHeBOCMVNBS plasmid was constructed as described (31). FADUNBS cell lines were generated by transfecting the HeBOCMVNBS plasmid into FADU cells and selection under G418 (200 μg/mL). Vector control cell lines (FADUCMV) were generated by transfecting pHeBOCMV into FADU cells.

The plasmids pSUPERIORNBSi and pSUPERIORNBSim were generated by inserting the oligonucleotide of 5′-GATCCCCTTCAGAATAGAGTATGAGCCTTCAAGAGAGGCTCATACTCTATTCTGAATTTTTGGAAA-3′ and the mismatched oligonucleotide of 5′-GATCCCCTTCAGAATATAGTATGAGCCTTCAAGAGAGGCTCATACTATATTCTGAATTTTTGGAAA-3′ into the pSUPERIOR plasmid, respectively (OligoEngine, Inc., Seattle, WA). To establish the inducible FADUNBSi or FADUNBSim clones, the pcDNA6/TR was transfected into FADU cells and selected under blasticidin (5 μg/mL). After selection of the clones expressing Tet repressor (FADUTetR), the pSUPERIORNBSi or pSUPERIORNBSim plasmid was transfected into FADUTetR cells and selected under puromycin (0.25 μg/mL). The repression of endogenous NBS1 expression was induced by adding doxycycline (1 μg/mL) to FADUNBSi or FADUNBSim cells and incubated for 24 hours.

Soft agar clonogenicity assay. Anchorage-independent growth of FADU, FADUNBS, and FADUNBSi cells was examined by survival of colonies on soft agar as described previously, except that 15% FBS was used (36). Three different cell numbers (5 × 103, 104, and 2 × 104) were used. The dishes were incubated for 2 weeks and colonies were counted.

PI3K assay. PI3K assays were done as described (37). Briefly, the immunoprecipitate of FADUCMV, FADUNBS, FADUNBSi, or FADUNBSim extracts with an anti-p110α antibody (Santa Cruz Biotechnology) was washed thrice with TNTG buffer [20 mmol/L Tris (pH 7.5), 150 mmol/L NaCl, 0.1% Triton X-100, 10% glycerol], washed twice with PI3K reaction buffer [20 mmol/L Tris-HCl (pH 7.4), 100 mmol/L NaCl, 0.5 mmol/L EGTA], and suspended in 50 μL PI3K reaction buffer containing 0.2 mg phosphatidylinositol (bovine liver; Avanti Polar Lipids, Inc., Alabaster, AL)/mL. The reactions were initiated by adding 5 μL MgCl2-ATP mixture (200 mmol/L MgCl2, 200 μmol/L ATP) containing 10 μCi [γ-32P]ATP to the reaction mixture and incubating the mixture at 25°C for 20 minutes. The reactions were terminated by adding 150 μL chloroform/methanol/11.6 N HCl (100:200:2). After addition of 120 μL chloroform to each sample, the organic phase was separated by centrifugation and washed twice with methanol/1 N HCl (1:1). After evaporation, the pellets were resuspended in 20 μL chloroform, spotted onto a silica gel plate, and developed in chloroform/methanol/28% ammonium hydroxide/water (43:38:5:7). The phosphorylated lipids were visualized by autoradiography. The intensity of phosphorylated lipids was measured by PhosphoImager. Kinase activity was measured as percentage of lipids becoming phosphorylated by PI3K. Data are representative of results from two experiments using different FADUCMV and FADUNBS clones or repeated in FADUNBSi/FADUNBSim clones.

Statistical analysis. Statistical Package of Social Sciences software version 12.0 (SPSS, Inc., Chicago, IL) was used for statistical analysis. The Kaplan-Meier estimate was used for survival analysis, and the log-rank test was selected to compare the cumulative survival durations in different patient groups. The Cox's proportional hazards model was applied in multivariate survival analysis to test independent prognostic factors. The level of statistical significance was set at 0.05 for all tests.

Increased NBS1 expression in tumor tissues of advanced HNSCC patients. To investigate whether NBS1 overexpression may contribute to the tumorigenesis of HNSCC, immunohistochemistry analysis of NBS1 expression in 81 pairs of tumor and neighboring normal tissues of advanced HNSCC patients was done. In the normal tissue samples taken from these patients, 43 (53%) samples did not express NBS1 protein (immunohistochemistry level 0; Fig. 1A) and 38 (47%) samples showed nucleus staining only (immunohistochemistry level +; Fig. 1C). No NBS1 cytoplasmic staining could be detected in all normal tissues. For NBS1 expression in tumor tissues, 32 (40%) samples showed NBS1 negative staining or nucleus staining only (immunohistochemistry level 0 or +; Fig. 1B and D), 12 (15%) samples showed both nucleus and weak cytoplasmic staining (immunohistochemistry level ++; Fig. 1E), and 37 (45%) samples showed strong nucleus and cytoplasmic staining (immunohistochemistry level +++; Fig. 1F). These results indicated that increased NBS1 expression (immunohistochemistry level +++) in tumor tissues could be found in a significant proportion (45%) of advanced HNSCC patient samples (Fig. 2A).

Fig. 2.

Increased NBS1 mRNA expression and protein levels in representative pairs of HNSCC patient samples. A, scoring of different levels of NBS1 immunohistochemistry staining in 81 locally advanced HNSCC patient samples and their statistics. B, quantitative real-time PCR analysis of 10 representative HNSCC cases with NBS1 immunohistochemistry level +++. TBP was used as an internal control. Relative NBS1 mRNA expression was calculated as the ratio of NBS1 versus TBP in tumor tissues (T) and the neighboring normal tissues (N). C, Western blot analysis of NBS1 protein levels in three representative pairs of HNSCC samples (NBS1 immunohistochemistry level +++) in tumor tissues and the neighboring normal tissues. γ-Tubulin was used as a control for protein loading.

Fig. 2.

Increased NBS1 mRNA expression and protein levels in representative pairs of HNSCC patient samples. A, scoring of different levels of NBS1 immunohistochemistry staining in 81 locally advanced HNSCC patient samples and their statistics. B, quantitative real-time PCR analysis of 10 representative HNSCC cases with NBS1 immunohistochemistry level +++. TBP was used as an internal control. Relative NBS1 mRNA expression was calculated as the ratio of NBS1 versus TBP in tumor tissues (T) and the neighboring normal tissues (N). C, Western blot analysis of NBS1 protein levels in three representative pairs of HNSCC samples (NBS1 immunohistochemistry level +++) in tumor tissues and the neighboring normal tissues. γ-Tubulin was used as a control for protein loading.

Close modal

To confirm the immunohistochemistry results of NBS1 expression, real-time PCR and Western blot analysis were done in representative tissue samples (NBS1 immunohistochemistry level +++) to correlate immunohistochemistry staining results with mRNA expression and protein levels. About 1- to 5-fold increase in NBS1 mRNA expression was observed in tumor tissues compared with the neighboring normal tissues using real-time PCR analysis (Fig. 2B). There is no increase or <1-fold increase in the samples of NBS1 immunohistochemistry staining 0 to ++ (data not shown). In addition, increased NBS1 protein levels in tumor tissues of HNSCC were also shown in representative pairs of HNSCC patient samples by Western blot analysis (Fig. 2C). These results showed the increased NBS1 expression in a significant percentage (45%) of advanced HNSCC patient samples.

Clinical significance of increased NBS1 expression in advanced HNSCC patients. To investigate whether increased NBS1 expression levels are associated with clinical outcome, Kaplan-Meier survival analysis was done to determine the prognostic significance of different NBS1 immunohistochemistry staining levels. Patients with NBS1 immunohistochemistry level +++ staining were associated with a significantly shorter overall survival period compared with the immunohistochemistry level 0, +, and ++ group (Fig. 3A). We therefore categorized patients into two groups: NBS1 immunohistochemistry level +++ (37 cases, 45%) and NBS1 immunohistochemistry level 0 to ++ (44 cases, 55%). Survival analysis showed that patients in the NBS1 immunohistochemistry level +++ group had a significantly worse overall survival (P < 0.001; Fig. 3B). The 2-year overall survival rate was 37.9% for NBS1 immunohistochemistry level +++ patients and 73.2% for immunohistochemistry level 0 to ++ patients. Univariate survival analysis was also done in other pretreatment variables and revealed that young age and neck node involvement were also significant prognostic factors (P = 0.03 and 0.007, respectively; Table 2). To evaluate the independent prognostic effect of NBS1 overexpression, multivariate Cox proportional hazard model stratified survival analysis was done. Multivariate survival analysis was carried out for three variables for which there were significant prognostic factors in univariate analysis, including age, N status, and NBS1 expression. The results showed that both N status and increased NBS1 expression (immunohistochemistry level +++) were independent markers for predicting overall survival (P = 0.003 and 0.03, respectively; Table 3). To further confirm the prognostic effect of NBS1 overexpression in advanced HNSCC, we stratified patients into the N+ and N− group and found that increased NBS1 expression was a significant prognostic marker regardless of lymph node status (Fig. 3C and D). All the statistical analyses suggested that NBS1 overexpression is a significant prognostic marker and predictor of aggressive HNSCC.

Fig. 3.

Prognostic significance of increased NBS1 expression in advanced HNSCC patients. A, individual comparison of prognostic value between NBS1 immunohistochemistry level +++ versus level 0, +, and ++, respectively. Inset, P between each comparison. B, statistical comparison between NBS1 immunohistochemistry level +++ versus level 0 to ++ grouped together, which showed significantly shorter overall survival period in the NBS1 immunohistochemistry +++ patients (P < 0.001, log-rank). C and D, comparison of overall survival periods between NBS immunohistochemistry level +++ versus 0 to ++ in node-negative (N-; C) and node-positive (N+; D) HNSCC cases.

Fig. 3.

Prognostic significance of increased NBS1 expression in advanced HNSCC patients. A, individual comparison of prognostic value between NBS1 immunohistochemistry level +++ versus level 0, +, and ++, respectively. Inset, P between each comparison. B, statistical comparison between NBS1 immunohistochemistry level +++ versus level 0 to ++ grouped together, which showed significantly shorter overall survival period in the NBS1 immunohistochemistry +++ patients (P < 0.001, log-rank). C and D, comparison of overall survival periods between NBS immunohistochemistry level +++ versus 0 to ++ in node-negative (N-; C) and node-positive (N+; D) HNSCC cases.

Close modal
Table 2.

Univariate survival analysis

VariablesMedian overall survival (mo)P (log-rank)
Age   
    <50 20.4 0.030 
    ≥50 27.0  
Gender   
    Male 20.0 0.711 
    Female 26.0  
Primary tumor site   
    Hypopharynx 20.4 0.260 
    Larynx 22.4  
    Oropharynx 28.9  
    Oral cavity 26.0  
T stage   
    T3 25.0 0.471 
    T4 21.5  
N stage   
    N0 —* 0.007 
    N1-3 16.5  
NBS1 overexpression   
    Yes 15.4 <0.001 
    No —*  
VariablesMedian overall survival (mo)P (log-rank)
Age   
    <50 20.4 0.030 
    ≥50 27.0  
Gender   
    Male 20.0 0.711 
    Female 26.0  
Primary tumor site   
    Hypopharynx 20.4 0.260 
    Larynx 22.4  
    Oropharynx 28.9  
    Oral cavity 26.0  
T stage   
    T3 25.0 0.471 
    T4 21.5  
N stage   
    N0 —* 0.007 
    N1-3 16.5  
NBS1 overexpression   
    Yes 15.4 <0.001 
    No —*  
*

Median survival was not reached.

Table 3.

Multivariate survival analysis

VariablesHazard ratio (95% CI)P
Age (<50 vs ≥50) 1.81 (1.12-2.76) 0.121 
N stage (N1-3 vs N03.38 (1.56-6.11) 0.003 
NBS overexpression (yes vs no) 2.72 (1.39-3.52) 0.030 
VariablesHazard ratio (95% CI)P
Age (<50 vs ≥50) 1.81 (1.12-2.76) 0.121 
N stage (N1-3 vs N03.38 (1.56-6.11) 0.003 
NBS overexpression (yes vs no) 2.72 (1.39-3.52) 0.030 

NBS1 overexpression increased the transformation activity of HNSCC cells. To delineate the role of increased NBS1 expression in the tumorigenesis of HNSCC, the NBS1 expression vector (HeBOCMVNBS1; ref. 31) was stably transfected into a human hypopharyngeal cancer cell line (FADU) to establish NBS1-overexpressing clones (FADUNBS). Increased NBS1 expression could be identified in three FADUNBS stable clones compared with the vector (HeBOCMV)–transfected FADU clones (FADUCMV; Fig. 4A). Soft agar clonogenicity assay showed the significantly increased colony formation activity (2.5- to 3-fold of control) in the FADUNBS clones compared with the vector control FADUCMV clones (Fig. 4B). Figure 4C showed the representative picture of the FADUCMV versus the FADUNBS clone. Clonal populations of NBS1-overexpressing FADU cells also showed increased colony formation activity (>3-fold) compared with the vector control transfected FADU cells (data not shown). These results indicated that NBS1 overexpression increased the transformation activity of FADU cells. To further correlate NBS1 expression levels with the transformation activity of HNSCC cells, we constructed an inducible NBS1 knockdown FADUNBSi cell line in which endogenous NBS1 expression was repressed by doxycycline-inducible short interference RNA (siRNA) against NBS1 mRNA. Figure 4D showed that endogenous NBS1 expression of the FADUNBSi clone was successfully repressed (∼50% of preinduction levels) by inducible siRNA. Clonogenicity assay of FADUNBSi cells before and after doxycycline treatment showed the decreased transformation activity (∼55% of control) in the FADUNBSi clone following doxycycline induction (Fig. 4E). These results showed the correlation between NBS1 expression levels and transformation activity of FADU cells.

Fig. 4.

NBS1 overexpression increased the transformation activity of FADU cells. A, Western blot analysis showed the protein levels of NBS1 in the FADUCMV and FADUNBS clones. Actin was used as a control for protein loading. B, Soft agar colony formation assay was done in the FADUCMV and FADUNBS clones. C, a representative picture of clonogenicity assay (FADUCMV3 versus FADUNBS6) using 5 × 103 cells in each dish. D, Western blot analysis of the FADU vector control clone and the FADUNBSi clone following doxycycline (Dox) treatment to induce siRNA and repress endogenous NBS1 expression. E, soft agar colony formation assay in the FADU vector control clone and the FADUNBSi clone without or with doxycycline treatment. *, P < 0.05, statistical significance between without and with doxycycline treatment in the FADUNBSi clone.

Fig. 4.

NBS1 overexpression increased the transformation activity of FADU cells. A, Western blot analysis showed the protein levels of NBS1 in the FADUCMV and FADUNBS clones. Actin was used as a control for protein loading. B, Soft agar colony formation assay was done in the FADUCMV and FADUNBS clones. C, a representative picture of clonogenicity assay (FADUCMV3 versus FADUNBS6) using 5 × 103 cells in each dish. D, Western blot analysis of the FADU vector control clone and the FADUNBSi clone following doxycycline (Dox) treatment to induce siRNA and repress endogenous NBS1 expression. E, soft agar colony formation assay in the FADU vector control clone and the FADUNBSi clone without or with doxycycline treatment. *, P < 0.05, statistical significance between without and with doxycycline treatment in the FADUNBSi clone.

Close modal

Correlation between NBS1 overexpression and the activation of PI3K/Akt pathway in HNSCC cell lines and patient samples. Our recent results show that overexpression of NBS1 contributes to transformation through the activation of PI3K/Akt (32). To show whether a similar mechanism was present in HNSCC cells overexpressing NBS1, Western blot analysis was done. The results showed that Akt phosphorylation levels increased in the FADUNBS clones (Fig. 5A), and decreased Akt phosphorylation levels were observed in the FADUNBSi clone following doxycycline induction (Fig. 5B). To show the activation of Akt pathway by NBS1 overexpression, we screened different Akt downstream targets. Among them, mTOR, whose phosphorylation regulates protein synthesis to increase cell growth (38), was hyperphosphorylated in FADUNBS cells (Fig. 5A). Decreased mTOR phosphorylation levels were also identified in the FADUNBSi clone following doxycycline induction (Fig. 5B). The control experiment using inducible mismatched siRNA to repress endogenous NBS1 expression in FADUNBSim clones undergoing doxycycline induction showed no decrease in the levels of NBS1 expression and phosphorylated Akt (Fig. 5C), showing the specificity of siRNA in FADUNBSi clones following doxycycline induction. To test whether the activation of Akt in the FADUNBS clone was induced by increased upstream PI3K activity, we did the PI3K activity assay in FADUNBS versus FADUCMV cells as well as FADU control versus FADUNBSi/FADUNBSim cells before and after doxycycline induction. The result showed that the FADUNBS clone had a 2-fold increase in PI3K activity compared with the FADUCMV clone (Fig. 5D). Repression of endogenous NBS1 expression in FADUNBSi cells caused a 64% decrease in PI3K activity compared with no decrease in PI3K activity in FADUNBSim cells following doxycycline induction (Fig. 5E). These results showed that NBS1 overexpression correlated with the activation of PI3K/Akt pathway.

Fig. 5.

NBS1 overexpression correlated with the activation of PI3K/Akt pathway. A, Western blot analysis of the FADUCMV and FADUNBS clones showed the correlation between NBS1 expression and the phosphorylation levels of Akt (pAkt) and mTOR (p-mTOR). Total Akt and actin were used as protein loading controls. B, Western blot analysis showed the correlation between NBS1 expression and the phosphorylation levels of Akt and mTOR in the FADU vector control and FADUNBSi clone without or with doxycycline treatment. C, Western blot analysis showed the specific repression of endogenous NBS1 and phosphorylated Akt levels in FADUNBSi cells but not in FADUNBSim cells following doxycycline treatment. D, PI3K assay of the FADUCMV and FADUNBS clones. E, PI3K assay of the FADU control and FADUNBSim and FADUNBSi clones without or with doxycycline treatment.

Fig. 5.

NBS1 overexpression correlated with the activation of PI3K/Akt pathway. A, Western blot analysis of the FADUCMV and FADUNBS clones showed the correlation between NBS1 expression and the phosphorylation levels of Akt (pAkt) and mTOR (p-mTOR). Total Akt and actin were used as protein loading controls. B, Western blot analysis showed the correlation between NBS1 expression and the phosphorylation levels of Akt and mTOR in the FADU vector control and FADUNBSi clone without or with doxycycline treatment. C, Western blot analysis showed the specific repression of endogenous NBS1 and phosphorylated Akt levels in FADUNBSi cells but not in FADUNBSim cells following doxycycline treatment. D, PI3K assay of the FADUCMV and FADUNBS clones. E, PI3K assay of the FADU control and FADUNBSim and FADUNBSi clones without or with doxycycline treatment.

Close modal

To confirm that the activation of PI3K/Akt pathway by NBS1 overexpression could occur in HNSCC patient samples, immunohistochemistry staining of phosphorylated Akt was done in 20 NBS1 immunohistochemistry level +++ and 20 immunohistochemistry level 0 to ++ patient samples. Predominantly cytoplasmic staining of phosphorylated Akt (level ++ to +++ as defined in Materials and Methods; ref. 35) could be detected in 18 of 20 (90%) NBS1 immunohistochemistry level +++ patient samples (Fig. 6A and B). On the contrary, level 0 to + staining of phosphorylated Akt was shown in 20 of 20 (100%) NBS1 immunohistochemistry level 0 to ++ patient samples (Fig. 6C and D). Western blot analysis confirmed the activation of Akt and its downstream target mTOR in representative HNSCC patient samples (Fig. 6E). These results showed that activation of Akt by NBS1 overexpression indeed occurred in HNSCC patient samples.

Fig. 6.

Increased NBS1 expression correlated with the phosphorylation levels of Akt in HNSCC patient samples. A and B, immunohistochemistry staining of NBS1 and phosphorylated Akt showed the cytoplasmic colocalization of NBS1 (A) and phosphorylated Akt (B) expression in the NBS1 immunohistochemistry level +++ sample (white arrow). Black arrow, NBS1 nucleus staining. C and D, in the negative NBS1 staining case, there was neither NBS1 (C) nor phosphorylated Akt (D) staining. Magnification, ×400 (A-D). E, Western blot analysis of NBS1, phosphorylated Akt, and phosphorylated mTOR in two representative pairs of HNSCC patient samples (NBS1 immunohistochemistry level +++). Total Akt and γ-tubulin were used as protein loading controls.

Fig. 6.

Increased NBS1 expression correlated with the phosphorylation levels of Akt in HNSCC patient samples. A and B, immunohistochemistry staining of NBS1 and phosphorylated Akt showed the cytoplasmic colocalization of NBS1 (A) and phosphorylated Akt (B) expression in the NBS1 immunohistochemistry level +++ sample (white arrow). Black arrow, NBS1 nucleus staining. C and D, in the negative NBS1 staining case, there was neither NBS1 (C) nor phosphorylated Akt (D) staining. Magnification, ×400 (A-D). E, Western blot analysis of NBS1, phosphorylated Akt, and phosphorylated mTOR in two representative pairs of HNSCC patient samples (NBS1 immunohistochemistry level +++). Total Akt and γ-tubulin were used as protein loading controls.

Close modal

In this report, we showed that increased NBS1 expression (NBS1 immunohistochemistry level +++ staining) occurred in a significant proportion (45%) of advanced HNSCC patients and was a significant prognostic marker of advanced HNSCC. NBS1, as with many other DNA repair checkpoint proteins, was initially known to guard the integrity of the genome (1719). However, we propose that NBS1 may act as an oncoprotein and participate in the tumorigenesis of HNSCC due to the following reasons: (a) increased NBS1 expression is identified in 45% of advanced HNSCC patients and is associated with a poor prognosis; (b) overexpression of NBS1 increases the transformation activity of HNSCC cells, and siRNA approach to repress endogenous NBS1 expression decreases their transformation activity; and (c) NBS1 overexpression correlates with the activation of PI3K/Akt pathway. Our recent results showed that overexpression of NBS1 contributes to transformation through the activation of PI3K/Akt (32). Recent studies to identify prognostic markers in uveal melanoma also showed that NBS1 overexpression is a significant prognostic marker (39), which is consistent with the results of this report.

Increased phosphorylation levels of Akt and its downstream targets have been identified in different types of human cancer (38, 40, 41), and the PI3K/Akt pathway is regarded as one of the most important oncogenic pathways in human cancers (42). PI3K-initiated pathway is the most frequently altered in HNSCC (43) and activation of Akt is shown to be a frequent event in HNSCC (44). Overexpression of eIF4E with the activation of Akt/mTOR pathway was also noted in the surgical margins of HNSCC (45). Activation of Akt was constantly associated with disease progression in different types of cancer (35, 41, 44). In this article, increased Akt phosphorylation levels could be identified in most HNSCC patients with increased NBS1 expression (Fig. 6). The proliferation-inducing function of NBS1 is suggested by the expression of NBS1 in highly proliferating tissues developmentally (46) and by the ability of Mre11 complex to prevent double-strand break accumulation during chromosomal DNA synthesis to ensure cell cycle progression (20). In addition, the phenotypes of diminished expansion of the inner cell mass of mutant blastocysts (Nbs1-null) and cellular proliferation defects in Nbs1m/m mouse embryonic fibroblasts also supported the role of NBS1 in cellular proliferation (2830). Overexpression of NBS1 causes the activation of PI3K/Akt, which induces transformation through increase of cell proliferation, promotion of cell growth, and inhibition of apoptosis by phosphorylating its various downstream target proteins (mTOR in this article; ref. 38). Activation of PI3K/Akt by increased NBS1 expression argues against the contention that NBS1 overexpression is just an accompanying side event during HNSCC development. Preliminary results showed the coimmunoprecipitation of NBS1 and one of the PI3K subunits, suggesting their direct interaction,7

7

M.H. Yang and K.J. Wu, unpublished results.

which may lead to the activation of PI3K. Increased NBS1 expression may be due to the activation of c-MYC as we reported previously (31) or the involvement of other mechanisms because a low percentage (∼10%) of HNSCC showed c-MYC overexpression (13).

In conclusion, our results show that overexpression of the DNA double-strand break repair gene NBS1 is capable of contributing to transformation and tumorigenesis possibly through the activation of PI3K/Akt, thereby further elucidating the molecular mechanism of tumorigenesis in HNSCC. Increased NBS1 expression is an independent prognostic marker of tumor progression in advanced HNSCC. This discovery provides valuable information for the identification and characterization of a novel molecular marker and a target for future therapeutic purposes in HNSCC.

Grant support: Taipei Veterans General Hospital grant VGH 94-171 (M.H. Yang), National Health Research Institutes grant NHRI-EX94-9329SI (K.J. Wu), National Research Program for Genomic Medicine Department of Health grant DOH94-TD-G-111-012 (K.J. Wu), and National Science Council grants NSC93-2320-B-010-062 (K.J. Wu) and NSC94-2314-B-075-077 (M.H. Yang).

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.

We thank Dr. W.Y. Li (Department of Pathology, Taipei Veterans General Hospital) for providing expert opinions on pathology reading, Drs. H. Li and S.K. Tai for providing the pSUPERIOR plasmid and critical comment on the article, and C.Y. Chang for excellent technical assistance.

1
Hardisson D. Molecular pathogenesis of head and neck squamous cell carcinoma.
Eur Arch Otorhinolaryngol
2003
;
260
:
502
–8.
2
Department of Health, the Executive Yuan, Republic of China. Cancer Registry Annual Report in Taiwan area, 1999 to April 2003.
3
Ho PS, Ko YC, Yang YH, et al. The incidence of oropharyngeal cancer in Taiwan: an endemic betel quid chewing area.
J Oral Pathol Med
2002
;
31
:
213
–9.
4
Maier H, Dietz A, Gewelke U, et al. Tobacco and alcohol and the risk of head and neck cancer.
Clin Invest
1992
;
70
:
320
–7.
5
Induction chemotherapy plus radiation compared with surgery plus radiation in patients with advanced laryngeal cancer: the Department of Veterans Affairs Laryngeal Cancer Study Group.
N Engl J Med
1991
;
324
:
1685
–90.
6
Forastiere AA, Goepfert H, Maor M, et al. Concurrent chemotherapy and radiotherapy for organ preservation in advanced laryngeal cancer.
N Engl J Med
2003
;
349
:
2091
–8.
7
Chen PH, Ko YC, Yang YH, et al. Important prognostic factors of long-term oropharyngeal carcinoma survivors in Taiwan.
Oral Oncol
2004
;
40
:
847
–55.
8
Matsuo JM, Patel SG, Singh B, et al. Clinical nodal stage is an independently significant predictor of distant failure in patients with squamous cell carcinoma of the larynx.
Ann Surg
2003
;
238
:
412
–21.
9
Wendt TG, Bank P. Prognostic factors in squamous cell carcinoma of the head and neck.
Onkologie
2002
;
25
:
208
–12.
10
Perez CA, Patel MM, Chao KS, et al. Carcinoma of the tonsillar fossa: prognostic factors and long-term therapy outcome.
Int J Radiat Oncol Biol Phys
1998
;
42
:
1077
–84.
11
Lydiatt WM, Shah JP, Hoffman HT. American Joint Committee on Cancer. AJCC stage groupings for head and neck cancer: should we look at alternatives? A report of the Head and Neck Sites Task Force.
Head Neck
2001
;
23
:
607
–12.
12
Yarbrough WG, Shores C, Witsell DL, et al. ras mutations and expression in head and neck squamous cell carcinomas.
Laryngoscope
1994
;
104
:
1337
–47.
13
Freier K, Joos S, Flechtenmacher C, et al. Tissue microarray analysis reveals site-specific prevalence of oncogene amplification in head and neck squamous cell carcinoma.
Cancer Res
2003
;
63
:
1179
–82.
14
Ang KK, Berkey BA, Tu X, et al. Impact of epidermal growth factor receptor expression on survival and pattern of relapse in patients with advanced head and neck carcinoma.
Cancer Res
2002
;
62
:
7350
–6.
15
Xie X, Clausen OP, De Angelis P, et al. The prognostic value of spontaneous apoptosis, Bax, Bcl-2, and p53 in oral squamous cell carcinoma of the tongue.
Cancer
1999
;
86
:
913
–20.
16
Erber R, Conradt C, Homann N, et al. TP53 DNA contact mutations are selectively associated with allelic loss and have a strong clinical impact in head and neck cancer.
Oncogene
1998
;
16
:
1671
–9.
17
Petrini JHJ. The Mre11 complex and ATM: collaborating to navigate S phase.
Curr Opin Cell Biol
2000
;
12
:
293
–6.
18
Karran P. DNA double strand break repair in mammalian cells.
Curr Opin Genet Dev
2000
;
10
:
144
–50.
19
D'Amours D, Jackson SP. The Mre11 complex: at the crossroads of DNA repair and checkpoint signaling.
Nat Rev Mol Cell Biol
2002
;
3
:
317
–27.
20
Costanzo V, Robertson K, Bibikova M, et al. Mre11 protein complex prevents double-strand break accumulation during chromosomal DNA replication.
Mol Cell
2001
;
8
:
137
–47.
21
Lim DS, Kim ST, Xu B, et al. ATM phosphorylates p95/nbs1 in an S-phase checkpoint pathway.
Nature
2000
;
404
:
613
–7.
22
Zhao S, Weng YC, Yuan SS, et al. Functional link between ataxia-telangiectasia and Nijmegen breakage syndrome gene products.
Nature
2000
;
405
:
474
–7.
23
Wu X, Ranganathan V, Weisman DS, et al. ATM phosphorylation of Nijmegen breakage syndrome protein is required in a DNA damage response.
Nature
2000
;
405
:
477
–82.
24
Stumm M, von Ruskowsky A, Siebert R, et al. No evidence for deletions of the NBS1 gene in lymphomas.
Cancer Genet Cytogenet
2001
;
26
:
60
–2.
25
Varon R, Reis A, Henze G, et al. Mutations in the Nijmegen breakage syndrome gene (NBS1) in childhood acute lymphoblastic leukemia (ALL).
Cancer Res
2001
;
61
:
3570
–2.
26
Plisiecka-Halasa J, Dansonka-Mieskowska A, Rembiszewska A, et al. Nijmegen breakage gene (NBS1) alterations and its protein (nibrin) expression in human ovarian tumors.
Ann Hum Genet
2002
;
66
:
353
–9.
27
Cerosaletti KM, Morrison VA, Sabath DE, et al. Mutations and molecular variants of the NBS1 gene in non-Hodgkin lymphoma.
Genes Chromosomes Cancer
2002
;
35
:
282
–6.
28
Zhu J, Petersen S, Tessarollo L, et al. Targeted disruption of the Nijmegen breakage syndrome gene NBS1 leads to early embryonic lethality in mice.
Curr Biol
2001
;
11
:
105
–9.
29
Williams BR, Mirzoeva OK, Morgan WF, et al. A murine model of Nijmegen breakage syndrome.
Curr Biol
2002
;
12
:
648
–53.
30
Kang J, Bronson RT, Xu Y. Targeted disruption of NBS1 reveals its roles in mouse development and DNA repair.
EMBO J
2002
;
21
:
1447
–55.
31
Chiang YC, Teng SC, Su YN, et al. c-MYC directly regulates the transcription of NBS1 gene involved in DNA double-strand break repair.
J Biol Chem
2003
;
278
:
19286
–91.
32
Chen YC, Su YN, Chou PC, et al. Overexpression of NBS1 contributes to transformation through the activation of phosphatidylinositol 3-kinase/Akt.
J Biol Chem
2005
;
280
:
32505
–11.
33
Therasse P, Arbuck SG, Eisenhauer EA, et al. New guidelines to evaluate the response to treatment in solid tumors.
J Natl Cancer Inst
2000
;
92
:
205
–16.
34
Angele S, Treilleux I, Bremond A, et al. Altered expression of DNA double-strand break detection and repair proteins in breast carcinomas.
Histopathology
2003
;
43
:
347
–53.
35
David O, Jett J, LeBeau H, et al. Phospho-Akt overexpression in non-small cell lung cancer confers significant stage-independent survival disadvantage.
Clin Cancer Res
2004
;
10
:
6865
–71.
36
Wu KJ, Polack A, Dalla-Favera R. Coordinated regulation of iron-controlling genes, H-ferritin and IRP2, by c-MYC.
Science
1999
;
283
:
676
–9.
37
Ueki K, Algenstaedt P, Mauvais-Jarvis F. Positive and negative regulation of phosphoinositide 3-kinse-dependent signaling pathways by three different gene products of the p85 regulatory subunit.
Mol Cell Biol
2000
;
20
:
8035
–46.
38
Vivanco I, Sawyers CL. The phosphatidylinositol 3-kinase-Akt pathway in human cancer.
Nat Rev Cancer
2002
;
2
:
489
–501.
39
Ehlers JP, Harbour JW. NBS1 expression as a prognostic marker in uveal melanoma.
Clin Cancer Res
2005
;
11
:
1849
–53.
40
Staal SP. Molecular cloning of the akt oncogene and its human homologues AKT1 and AKT2: amplification of AKT1 in a primary human gastric adenocarcinoma.
Proc Natl Acad Sci U S A
1987
;
84
:
5034
–7.
41
Zhou X, Tan M, Stone Hawthorne V, et al. Activation of the Akt/mammalian target of rapamycin/4E-BP1 pathway by ErbB2 overexpression predicts tumor progression in breast cancers.
Clin Cancer Res
2004
;
10
:
6779
–88.
42
Testa JR, Bellacosa A. AKT plays a central role in tumorigenesis.
Proc Natl Acad Sci U S A
2001
;
98
:
10983
–5.
43
Pedrero JM, Carracedo DG, Pinto CM, et al. Frequent genetic and biochemical alterations of the PI 3-kinase/Akt/PTEN pathway in head and neck squamous cell carcinoma.
Int J Cancer
2005
;
114
:
242
–8.
44
Amornphimoltham P, Sriuranpong V, Patel V, et al. Persistent activation of the Akt pathway in head and neck squamous cell carcinoma: a potential target for UCN-01.
Clin Cancer Res
2004
;
10
:
4029
–37.
45
Nathan CA, Amirghahari N, Abreo F, et al. Overexpressed eIF4E is functionally active in surgical margins of head and neck cancer patients via activation of the Akt/mammalian target of rapamycin pathway.
Clin Cancer Res
2004
;
10
:
5820
–7.
46
Wilda M, Demuth I, Concannon P, Sperling K, Hameister H. Expression pattern of the Nijmegen breakage syndrome gene, Nbs1, during murine development.
Hum Mol Genet
2000
;
9
:
1739
–44.