Human papilloma viruses (HPV) are linked to an epidemic increase in oropharyngeal head and neck squamous cell carcinomas (HNSCC), which display viral inactivation of tumor suppressors TP53 and RB1 and rapid regional spread. However, the role of genomic alterations in enabling the modulation of pathways that promote the aggressive phenotype of these cancers is unclear. Recently, a subset of HPV+ HNSCC has been shown to harbor novel genetic defects or decreased expression of TNF receptor–associated factor 3 (TRAF3). TRAF3 has been implicated as a negative regulator of alternative NF-κB pathway activation and activator of antiviral type I IFN response to other DNA viruses. How TRAF3 alterations affect pathogenesis of HPV+ HNSCC has not been extensively investigated. Here, we report that TRAF3-deficient HPV+ tumors and cell lines exhibit increased expression of alternative NF-κB pathway components and transcription factors NF-κB2/RELB. Overexpression of TRAF3 in HPV+ cell lines with decreased endogenous TRAF3 inhibited NF-κB2/RELB expression, nuclear localization, and NF-κB reporter activity, while increasing the expression of IFNA1 mRNA and protein and sensitizing cells to its growth inhibition. Overexpression of TRAF3 also enhanced TP53 and RB tumor suppressor proteins and decreased HPV E6 oncoprotein in HPV+ cells. Correspondingly, TRAF3 inhibited cell growth, colony formation, migration, and resistance to TNFα and cisplatin-induced cell death. Conversely, TRAF3 knockout enhanced colony formation and proliferation of an HPV+ HNSCC line expressing higher TRAF3 levels. Together, these findings support a functional role of TRAF3 as a tumor suppressor modulating established cancer hallmarks in HPV+ HNSCC.

Significance: These findings report the functional role of TRAF3 as a tumor suppressor that modulates the malignant phenotype of HPV+ head and neck cancers. Cancer Res; 78(16); 4613–26. ©2018 AACR.

Head and neck squamous cell carcinoma (HNSCC) is the sixth most common cancer, with an annual incidence of 650,000 new cases and ∼200,000 deaths worldwide (1, 2). Persistent infection with high-risk human papillomavirus subtypes HPV16 and HPV18 has been established as an important risk factor for HNSCC that develop predominantly in the oropharyngeal tonsils (3). Since 1981, there has been a notable 225% increase in HPV+ HNSCC, while the incidence of smoking-related HPV HNSCC has declined (4, 5). Clinically, the HPV+ subset exhibits better responses to therapies and survival rates than similarly advanced HPV tumors. However, HPV+ HNSCC are distinguished by aggressive spread and growth within regional lymph nodes, which require major surgery or toxic chemoradiotherapy regimens (2, 3). The factors that contribute to the molecular pathogenesis of these unique features of HPV+ HNSCC remain incomplete.

HPV16 and 18 carry early genes E6 and E7 encoding oncoproteins that target key pathways, deregulating host resistance to infection and cellular proliferation, to promote the viral life cycle. HPV E6 expression in keratinocytes can repress type-I IFN and promote proliferative genes, to enhance viral protein synthesis and proliferation of virally infected cells (6). Further studies have shown that HPV infection can induce ubiquitin carboxyl-terminal hydrolase L1 (UCHL1), which can inhibit K63 ubiquination, important in Tank binding kinase-immune response factor 3 (TBK-IRF3)-mediated type-I IFN expression (7). HPV E6 can also commandeer and activate the so-called alternative nuclear factor-κB2 (NF-κB2) transcription factors and antiapoptotic genes, which promote resistance of keratinocytes to TNF, an important mediator of antiviral immunity (8). Critically, the HPV E6 and E7 oncoproteins also strategically target for degradation the tumor suppressor proteins TP53 and RB, which control the cell cycle (9). Interestingly, however, few individuals exposed to HPV develop chronic infection and HNSCC. These observations suggest that additional genetic alteration(s) and host factors may also affect how HPV mediates suppression of IFNs, NF-κB activation, inhibition of TP53 and RB gene expression, and the malignant phenotype.

Recently, we and The Cancer Genome Atlas (TCGA) Network uncovered a subset of HPV+ HNSCCs that harbor deletions of the chromosome region 14q32.32, deleterious truncating mutations, and/or decreased expression, affecting the gene TNF receptor–associated factor 3 (TRAF3; ref. 10). These deletions were not due to viral integration and disruption of the TRAF3 gene. Intriguingly, TRAF3 is a unique adaptor protein and ubiquitin ligase implicated as a negative regulator of the aforementioned alternative NF-κB2/RELB pathway (11). TRAF3 promotes cIAP-mediated ubiquitination and proteasome-dependent degradation of the pivotal NF-κB inducing kinase (NIK) protein, which mediates signal activation of the alternative pathway. Lymphotoxin-β (LTβ) and other ligands, which are richly expressed in the oropharyngeal tonsils and lymph nodes where HPV+ HNSCC arise and spread, bind receptors to activate NIK, IKKα, processing of NF-κB2 precursor p100 to p52, and nuclear translocation of transcriptionally active NF–κB2–p52/RELB dimers. Attenuation of TRAF3 has previously been implicated in the transcription of genes affecting cell fate, proliferation, and survival of lymphoid cells and hematopoietic malignancies (11, 12). Strikingly, TRAF3 has also been shown to serve a dual function in interferon responses to other DNA viruses (11, 13). However, the functional role of genetic alterations or reduced expression of TRAF3 in modulating alternative NF-κB pathway activation, IFN expression, repression of tumor suppressors TP53 and RB, and the malignant phenotype in HPV+ HNSCC has not been established.

In this study, we examined and revealed a novel role for decreased TRAF3 in fostering deregulation of these pathways and promoting pathogenesis of a subset of HPV+ HNSCC. These findings provide new opportunities for basic and clinical research that may lead to better diagnosis, prevention, and treatment of HPV+ HNSCC.

HNSCC patient samples and TCGA bioinformatics analysis

TCGA Project Management has collected necessary human subjects documentation to ensure that the project complies with 45-CFR-46 (the “Common Rule”). The program has obtained documentation from every contributing clinical site to verify that Institutional Review Board approval and informed consent has been obtained to participate in TCGA. The characteristics and data repositories for 279 HNSCC specimens and 16 normal mucosa were previously reported, including 36 HPV+, 243 HPV HNSCC, and 16 normal samples (10). Exome-wide sequencing was performed on all samples, and normalized genomic data for 279 HNSCC and 16 normal samples displaying a squamous keratin gene signature was obtained from the TCGA Genome Data Analysis Center and downloaded on September 19, 2012. All genomic data were processed through standard TCGA analytic pipelines and accessible through TCGA data portals. Significant focal copy-number alterations were extracted from the GISTIC2.0 processing pipeline, and data aggregated on significantly altered lesions are plotted by false discovery rate (FDR) less than 5% and P values of less than 0.05. Differential gene expression analysis of tumor versus normal samples was performed using DeSeq, and all data were log2 transformed (Bioconductor version 2.12). Molecular HPV signatures were identified using gene expression and somatic substitutions. Gene set enrichment analysis was performed using the online Cancer Genomics cBioportal database (http://www.cbioportal.org/).

HNSCC lines, cell culture, genomic DNA extraction, and PCR

UMSCC [University of Michigan (Ann Arbor, MI) series of HNSCC], UPCI (University of Pittsburgh Cancer Institute, Pittsburgh, PA), and VU (Free University Amsterdam, Amsterdam, the Netherlands) cell lines and clinical information were provided by Drs. Thomas E. Carey, Mark E. Prince, Carol R. Bradford (University of Michigan), Robert Ferris, and Susanne Gollin (University of Pittsburgh), or from previous publications (Supplementary Table S1; refs. 14–16). Cell authentication of UMSCC, UPCI, and VU lines was done at the University of Michigan by DNA genotyping of alleles for 9 loci (D3S1358, D5S818, D7S820, D8S1179, D13S317, D18S51, D21S11, FGA, vWA) and the amelogenin locus as described previously (15). A panel of 8 HPV-positive (UMSCC 47, 104, 2, 90, 152, 154, 93VU147T, and 105) and 8 HPV-negative (UMSCC 1, 9, 11A, 11B, 38, 46, 74A, and 74B) HNSCC cell lines was selected for evaluation of HPV type and TRAF3 expression. These cell lines were maintained in MEM or DMEM with 10% fetal calf serum (Life Technologies) at 37°C with 5% CO2 for a maximum of 8 weeks. Primary human oral keratinocytes (HOK) were cultured, in accordance with the supplier's protocol (Science Cell Research). Prior to experiments, all cells were confirmed to be Mycoplasma negative (by MycoAlert kit, cat. no. M7006, Thermo Fisher). Genomic DNA was extracted using a DNeasy Blood and Tissue Kit (cat. no. 51104, Qiagen) from selected frozen cell pellets. These cell lines were further evaluated for HPV status by PCR (Human Papillomavirus Detection Set, cat. no. 6602, Takara Bio).

RNA-seq analysis

Total RNA of 3 primary human keratinocyte and individual HNSCC cell line was isolated by combination of TRIzol (cat no. 15-596-026, Life Technologies) and QIAGEN RNeasy Mini Kit procedure (cat. no. 74104, QIAGEN). Ribosomal RNA was depleted using the Ribo-Zero kit (cat no. MRZH11124, Epicentre). Multiplexed whole transcriptome libraries were generated by SOLiD Total RNA-Seq Kit and SOLiD RNA Barcoding Kit (cat. no. 4427046, Life Technologies), and fragmented cDNA libraries were clonally amplified by emulsion PCR. The multiplex libraries were sequenced utilizing 75 bp forward and 35 bp reverse paired-end sequencing chemistry on the ABI SOLiD system. Reads were mapped into human NCBI Build 37 reference genome (Hg19) using LifeScope v.2.5 Genomic Analysis software (https://www.appliedbiosystems.com/lifescope). The read count of the genes was normalized using DESeq (estimateSizeFactors) R package (version 3.2.0) and upper quartile normalization method. Expression analysis was performed by comparison with normal HOK cell line expression, and gene expression was quantified using RSEM (RNA-Seq by Expectation Maximization).

Plasmids, siRNA, and transduction

YFP-pTo-TRAF3 fusion expression vector was kindly provided by Dr. Christina M. Annunziata, NCI/NIH, Bethesda, MD (16). His-tagged TRAF3 wild-type was generated by GeneCopoeia. For efficient retroviral transductions, certain cell lines were engineered to express these indicated vectors. SMART pool siRNAs targeting human TRAF3 (cat. no. E-005252) and nontargeting siRNA control (M-HUMAN-XX-0005) were purchased from Dharmacon. Cell transfection was carried out using X-tremeGENE HP DNA Transfection Reagent or X-TremeGENE siRNA Transfection Reagent (cat. no. 10465500, 063662, Roche), following the manufacturer's instructions. Stably transfected cell lines were allowed to recover for 48 hours before antibiotic selection, and only confirmed surviving pools were utilized for subsequent analysis.

TRAF3 CRISPR knockout cells

UPCI-SCC-90 and UMSCC47 cells were cotransfected with TRAF3 CRISPR/Cas9 KO and TRAF3 HDR plasmids (Santa Cruz Biotechnology) at equivalent ratios using lipofectamine 2000 (cat. no. 11668019, Thermo Fisher). Forty-eight hours after transfection, the cells were plated at low density with the addition of 1 μg/mL puromycin (cat. no. A1113802, Thermo Fisher) for selection of stable clones. Successful knockdown was confirmed with RT-PCR. Clonogenic assay was performed as described (17).

Statistical analysis

Data are shown as means ± SEM. Student t test or two-way ANOVA were performed using GraphPad. Statistical analysis is specifically indicated for each experiment (*, P < 0.05; **, P < 0.01; ***, P < 0.001; ****, P < 0.0001).

Additional methods of preparation of total RNA isolation, cDNA synthesis, qRT-PCR analysis, protein extracts, Western blot analysis, reporter assay, immunofluorescence staining, treatments, and cell viability are presented in the Supplementary Information.

TRAF3 alterations are associated with HPV+ HNSCC status

The recent TCGA study revealed that a subset of HPV+ HNSCC display novel recurrent deletions, truncating mutations, and/or decreased expression of TRAF3 (10). Here, we further show that TRAF3 focal or broader deletions of chromosome 14 (Chr14q32.32) are mainly observed in HPV+ HNSCC (Fig. 1A). Among 36 HPV+ tumors in this dataset, 14% homozygous and 25% heterozygous deletions were found, whereas in 243 HPV tumors, only 1.2% homozygous and 10% heterozygous deletions were observed (Fig. 1B, Fisher exact test, P = 2.1 × 10−6). In contrast, 38% of HPV tumors exhibited TRAF3 copy-number gains compared with only 6% of HPV+ tumors. Together, the different distribution of these DNA copy-number variations were associated with TRAF3 mRNA expression in HPV+ (P = 3.52 × 10−5) and HPV HNSCC samples (P = 2.0 × 1010; Fig. 1C). Furthermore, three cases with deleterious frameshift or nonsense mutations were observed only in HPV+ HNSCC.

Figure 1.

TRAF3 deletion is associated with HPV+ HNSCC status. A, Copy number variations (CNV) were analyzed and data were extracted from TCGA HNSCC project. TRAF3 chromosome location and copy number variation were presented on the Chr14q32.32 segmentation map for 36 HPV+ (top) and 243 HPV (bottom) HNSCC tumor specimens. The blue bar at the top right corner of chromosome represents the Chr14q32.32 region where TRAF3 is located. The color gradients depict the extent of inferred copy number loss (blue) and amplifications (red), respectively. The magnified view (marked by the black box) refers to sorted HPV+ HNSCC tumors according to the loss of TRAF3 locus (indicated by yellow lines at the bottom) and neighboring gene regions. B, Percentage of HPV+ (left) or HPV (right) HNSCC specimens with TRAF3 copy number variations. Copy number variation is presented on the x-axis and divided into five categories by GISTIC: homozygous deletion, −2; heterozygous deletion, −1; diploid, 0; one copy gain, 1; two copy amplification, 2. Significance of association between TRAF3 loss and HPV status was observed (Fisher exact test, P = 2.12 × 106). C, The significant association between TRAF3 expression (log2 RSEM; y-axis) and copy number variation (x-axis) was observed in both HPV+ (left) and HPV HNSCC after assessment by Pearson correlation test. The box plot indicates 25th, 50th, and 75th percentiles of TRAF3 gene expression. The whiskers mark the minimal and maximum values, excluding the outliers. The red triangle and diamond represent frameshift and nonsense mutations. D, TRAF3 mRNA expression of selected HPV+ and HPV HNSCC cell lines compared with HOK cells (control). *, P < 0.05 by Student t test. E, Whole cell lysates were harvested, and TRAF3 protein levels were examined by Western blot for HPV+ HNSCC cell lines. The histogram of relative protein expression was generated from densitometry analysis of relevant protein bands after adjustment to loading control of β-actin and normalized to HOK cells (control).

Figure 1.

TRAF3 deletion is associated with HPV+ HNSCC status. A, Copy number variations (CNV) were analyzed and data were extracted from TCGA HNSCC project. TRAF3 chromosome location and copy number variation were presented on the Chr14q32.32 segmentation map for 36 HPV+ (top) and 243 HPV (bottom) HNSCC tumor specimens. The blue bar at the top right corner of chromosome represents the Chr14q32.32 region where TRAF3 is located. The color gradients depict the extent of inferred copy number loss (blue) and amplifications (red), respectively. The magnified view (marked by the black box) refers to sorted HPV+ HNSCC tumors according to the loss of TRAF3 locus (indicated by yellow lines at the bottom) and neighboring gene regions. B, Percentage of HPV+ (left) or HPV (right) HNSCC specimens with TRAF3 copy number variations. Copy number variation is presented on the x-axis and divided into five categories by GISTIC: homozygous deletion, −2; heterozygous deletion, −1; diploid, 0; one copy gain, 1; two copy amplification, 2. Significance of association between TRAF3 loss and HPV status was observed (Fisher exact test, P = 2.12 × 106). C, The significant association between TRAF3 expression (log2 RSEM; y-axis) and copy number variation (x-axis) was observed in both HPV+ (left) and HPV HNSCC after assessment by Pearson correlation test. The box plot indicates 25th, 50th, and 75th percentiles of TRAF3 gene expression. The whiskers mark the minimal and maximum values, excluding the outliers. The red triangle and diamond represent frameshift and nonsense mutations. D, TRAF3 mRNA expression of selected HPV+ and HPV HNSCC cell lines compared with HOK cells (control). *, P < 0.05 by Student t test. E, Whole cell lysates were harvested, and TRAF3 protein levels were examined by Western blot for HPV+ HNSCC cell lines. The histogram of relative protein expression was generated from densitometry analysis of relevant protein bands after adjustment to loading control of β-actin and normalized to HOK cells (control).

Close modal

To determine if there are HPV+ HNSCC cell lines with decreased TRAF3 expression suitable for study, we investigated TRAF3 expression in a panel of eight HPV+ and HPV HNSCC cell lines (Supplementary Table S1), using RNA sequencing data. Overall, we observed significantly increased TRAF3 mRNA expression in HPV, compared with normal HOK and HPV+ cell lines (P < 0.05, Fig. 1D), consistent with the few HPV+ tumors and cell lines with low expression, as well as high number of HPV tumors and cell lines with gain of expression of TRAF3. We further compared TRAF3 protein levels in the 8 HPV+ HNSCC cell lines with normal human oral keratinocytes (HOK). UMSCC47 and UMSCC104 cell lines exhibited lower TRAF3 protein levels as assessed by Western blot (Fig. 1E), and these lines were selected for further study of the effects of decreased TRAF3 expression and modulation.

Decreased TRAF3 is associated with increased expression of alternative NF-κB pathway components in HPV+ HNSCC tumors

As TRAF3 was previously identified as a negative regulator of the alternative NF-κB pathway in immune and hematopoietic malignant cells (11, 12), we asked whether altered TRAF3 is associated with NF-κB pathway alterations in HNSCC tissues. Using the HNSCC TCGA datasets, we identified an apparent co-occurrence between TRAF3 genomic and expression alterations with the mRNA expression of alternative NF-κB pathway components (Fig. 2A left, top), but not with those in classical NF-κB pathway (Fig. 2A left, bottom), in HPV+ HNSCC tumors. Conversely, no such co-occurrence was observed in HPV HNSCC tumors (Fig. 2A, right). Supporting this, we observed a significantly higher average percentage of alterations in the NF-κB alternative pathway in HPV+ tumors, compared with HPV tumors, whereas no significant difference was observed in the classic NF-κB pathway (Fig. 2B). Finally, the co-occurrence and mutual exclusivity of paired alterations were calculated, and the relationships with statistical significance are indicated as color intensities (Fig. 2C; Supplementary Table S2). We observed more frequent co-occurrence of alterations in the key components of the alternative than the classic NF-κB pathway in HPV+ HNSCC tumors (Fig. 2C, left). In contrast, fewer tumor samples with strong and intermediate co-occurrences were observed in HPV HNSCC (Fig. 2C, right). These analyses indicate that deficient TRAF3 is associated with increased expression of alternative NF-κB pathway components in HPV+ HNSCC tumor specimens.

Figure 2.

TRAF3 deletion is associated with alternative NF-κB activation in HPV+ HNSCC tissues. A, Genomic alteration profiles of the alternative (top) and classic (bottom) NF-κB signaling pathway members across the TCGA dataset are presented by oncoprint, composed of HPV+ (left) and HPV (right) HNSCC samples. Each row represents a gene and each column represents a tumor sample, respectively. The percentage of alteration for individual gene was calculated by cBioPortal. B, Histograms representing the mean percentage of alterations for alternative (top) or classic (bottom) NF-κB signaling pathway members as presented above, respectively. ***, P < 0.001; ns, not significant. C, Heat maps summarizing co-occurrence or exclusivity association of paired NF-κB pathway gene members in HPV+ (left) and HPV (right) HNSCC. The association was presented as log odds ratio, and the co-occurrence or exclusivity is depicted in blue or orange, respectively. P value was calculated using Fisher exact test; *, P < 0.05; **, P < 0.01; ***, P < 0.001.

Figure 2.

TRAF3 deletion is associated with alternative NF-κB activation in HPV+ HNSCC tissues. A, Genomic alteration profiles of the alternative (top) and classic (bottom) NF-κB signaling pathway members across the TCGA dataset are presented by oncoprint, composed of HPV+ (left) and HPV (right) HNSCC samples. Each row represents a gene and each column represents a tumor sample, respectively. The percentage of alteration for individual gene was calculated by cBioPortal. B, Histograms representing the mean percentage of alterations for alternative (top) or classic (bottom) NF-κB signaling pathway members as presented above, respectively. ***, P < 0.001; ns, not significant. C, Heat maps summarizing co-occurrence or exclusivity association of paired NF-κB pathway gene members in HPV+ (left) and HPV (right) HNSCC. The association was presented as log odds ratio, and the co-occurrence or exclusivity is depicted in blue or orange, respectively. P value was calculated using Fisher exact test; *, P < 0.05; **, P < 0.01; ***, P < 0.001.

Close modal

TRAF3 expression inhibits the alternative but not classic NF-κB signaling in HPV+ HNSCC cell lines

To test the hypothesis that decreased TRAF3 is permissive for activation of the alternative NF-κB pathway in HPV+ HNSCC, we examined the expression of alternative pathway components and the effects of transiently expressing His-tagged TRAF3 (His-TRAF3) or control (His-Control) vectors in the HPV+ UMSCC47 and UMSCC104 cells that displayed relatively lower endogenous TRAF3 expression. TRAF3 mRNA was significantly increased in both cell lines after TRAF3 transfection (Fig. 3A, top). Next, we evaluated the mRNA expression levels of key alternative pathway subunits RELB and NF-κB2, which are modulated at transcriptional and posttranslational levels, as well as classic (RELA/p65) NF-κB subunit, which is constitutively expressed and modulated posttranslationally (18). Consistent with this, transient TRAF3 overexpression in both HPV+ cell lines resulted in a significant reduction of RELB and NF-κB2 mRNA expression, without affecting RELA expression (Fig. 3A). We then evaluated the effect of TRAF3 expression on protein levels of key alternative NF-κB pathway components in the same cell lines and an additional HPV+ HNSCC cell line, 93VU174T, following TRAF3 transfection (Fig. 3B; Supplementary Fig. S1A). TRAF3 expression significantly reduced protein levels of components involved in the alternative NF-κB pathway, including its co-factor and ubiquitinase cIAP1 together with its substrate NIK, and processed NF-κB2/p52 and RELB subunits (Fig. 3B and C; Supplementary Fig. S1A). We observed comparatively smaller decreases in TRAF3-cIAP co-factor TRAF2 or alternate pathway kinase IKKα that mediates NF-κB2-p52/RELB (Supplementary Fig. S1B). Taken together, these results indicate that increasing expression of TRAF3 in HPV+ HNSCC cells with lower TRAF3 can suppress mRNA and/or protein levels of components of the alternative NF-κB pathway.

Figure 3.

TRAF3 downregulates mRNA and protein expression of molecules involved in the alternative NF-κB pathway in HPV+ HNSCC cells. A, HPV+ HNSCC cell lines UMSCC47 (left) and UMSCC104 (right) were transiently transfected with TRAF3 (His-TRAF3) or control (His-Control) plasmid for 24 hours, and mRNA expression of TRAF3 and alternative RELB, NF-κB2, and classic RELA subunits was examined by qRT-PCR. mRNA expression is represented relative to control as mean ± SD (from 6 replicates). *, P < 0.05; **, P < 0.01; ***, P < 0.001, Student t test. B, HPV+ UMSCC47 and UMSCC104 cells were transiently transfected with TRAF3 expression vector and whole-cell lysates were harvested 48 hours after transfection. Protein expression of TRAF3 and alternative NF-κB pathway components were examined by Western blots. C, Histograms for expression of indicated proteins were derived from densitometry analysis of bands after adjustment to loading control of β-actin and are presented relative to the cells transfected with control plasmid at 48 hours. Data, mean ± SEM from independent experiments with three HPV+ cell lines (UMSCC47, 104, and 93VU174T; Fig. 3B and Supplementary Fig. S1). Significant differences between TRAF3-transfected samples and controls refer to *, P < 0.05;**, P < 0.01; ***, P < 0.001.

Figure 3.

TRAF3 downregulates mRNA and protein expression of molecules involved in the alternative NF-κB pathway in HPV+ HNSCC cells. A, HPV+ HNSCC cell lines UMSCC47 (left) and UMSCC104 (right) were transiently transfected with TRAF3 (His-TRAF3) or control (His-Control) plasmid for 24 hours, and mRNA expression of TRAF3 and alternative RELB, NF-κB2, and classic RELA subunits was examined by qRT-PCR. mRNA expression is represented relative to control as mean ± SD (from 6 replicates). *, P < 0.05; **, P < 0.01; ***, P < 0.001, Student t test. B, HPV+ UMSCC47 and UMSCC104 cells were transiently transfected with TRAF3 expression vector and whole-cell lysates were harvested 48 hours after transfection. Protein expression of TRAF3 and alternative NF-κB pathway components were examined by Western blots. C, Histograms for expression of indicated proteins were derived from densitometry analysis of bands after adjustment to loading control of β-actin and are presented relative to the cells transfected with control plasmid at 48 hours. Data, mean ± SEM from independent experiments with three HPV+ cell lines (UMSCC47, 104, and 93VU174T; Fig. 3B and Supplementary Fig. S1). Significant differences between TRAF3-transfected samples and controls refer to *, P < 0.05;**, P < 0.01; ***, P < 0.001.

Close modal

TRAF3 expression modulates NF-κB2/p52 and RELB nuclear localization, and NF-κB activity in the HPV+ UMSCC47 cell line

To further characterize how TRAF3 modulates the cellular distribution of alternative RELB/NF-κB2 subunits in HPV+ HNSCC, we examined cytoplasmic and nuclear expression of these protein subunits by Western blot. Aberrant nuclear localization of RELB, NF-κB2/p52, as well as p100 was detected in control nuclear as well as cytoplasmic fractions (Fig. 4A), consistent with a prior report demonstrating enhanced nuclear expression of both p100/52 in HPV+ SCC and E6-expressing keratinocytes (19). Transfection of TRAF3 increased distribution of NF-κB2 p100/52 proteins in the cytoplasmic fraction while decreasing nuclear p100/p52 and RELB protein levels in independent experiments (Fig. 4A; Supplementary Fig. S2A and S2B). We further tested functional NF-κB activity by utilizing a NF-κB luciferase reporter gene assay in the UMSCC47 cell line. Consistent with the reduced nuclear RELB/p52 protein levels, we observed a significant reduction in functional NF-κB reporter activity in TRAF3 cotransfected UMSCC47 cells (Fig. 4B). Conversely, TRAF3 knockdown using three different siRNAs, significantly reduced TRAF3 mRNA expression in UMSCC47 cells (Fig. 4C). Consistent with reduced TRAF3 mRNA expression in Fig. 4C, we observed reduced TRAF3 and increased RELB protein by Western blot (Supplementary Fig. S2C), as well as enhanced nuclear RELB by immunofluorescence staining (Fig. 4D). TRAF3 knockdown was accompanied by increased NF-κB reporter activity in UMSCC47 cells (Fig. 4E). These results demonstrate that TRAF3 can inversely modulate alternative NF-κB pathway RELB and NF-κB2/p52 expression, cellular localization, and functional activity in HPV+ HNSCC cells.

Figure 4.

TRAF3 decreases nuclear NF-κB p52 and RELB localization, and NF-κB activity in HPV+ UMSCC47 cell line. A, UMSCC47 cells were transiently transfected with TRAF3 (GFP-TRAF3 pToCMV) or control (GFP-pToCMV) plasmids for 24 hours. Cytoplasmic and nuclear protein were harvested and assessed by Western blot. β-Actin or Lamin A was used as cytoplasmic or nuclear protein loading control, respectively. B, UMSCC47 cells were transiently transfected with TRAF3 or control expression vector with NF-κB reporter plus LacZ plasmids for 24 hours. NF-κB reporter activity was adjusted to β-galactosidase. C, TRAF3 mRNA expression decreases following TRAF3-specific siRNAs (siTRAF3a-c) at 24 hours after transfection. All depicted samples were normalized to scrambled control siRNA (si-Control) knockdown. Data, mean ± SEM derived from three replicates. D, Nuclear RELB localization by confocal immunofluorescence microscopy following TRAF3-specific siRNA (siTRAF3b) or scrambled control siRNA (si-Control) knockdown at 24 hours. Cells were fixed and stained with antibodies against TRAF3, RELB, and Alexa fluor–conjugated secondary antibodies. Representative images show the localization of TRAF3 (green), RELB (purple), DAPI (blue), and merged images. Scale bar, 10 μm. E, NF-κB–mediated luciferase activity increases following TRAF3-specific siRNA (siTRAF3a-c). Renilla luciferase assay was performed on the same lysates and was used to adjust the differences in transfection efficiencies. All depicted samples were normalized to scrambled si-Control knockdown. Data, mean ± SEM derived from 6 replicates. Significant differences between samples and controls refer to *, P < 0.05; **, P < 0.01.

Figure 4.

TRAF3 decreases nuclear NF-κB p52 and RELB localization, and NF-κB activity in HPV+ UMSCC47 cell line. A, UMSCC47 cells were transiently transfected with TRAF3 (GFP-TRAF3 pToCMV) or control (GFP-pToCMV) plasmids for 24 hours. Cytoplasmic and nuclear protein were harvested and assessed by Western blot. β-Actin or Lamin A was used as cytoplasmic or nuclear protein loading control, respectively. B, UMSCC47 cells were transiently transfected with TRAF3 or control expression vector with NF-κB reporter plus LacZ plasmids for 24 hours. NF-κB reporter activity was adjusted to β-galactosidase. C, TRAF3 mRNA expression decreases following TRAF3-specific siRNAs (siTRAF3a-c) at 24 hours after transfection. All depicted samples were normalized to scrambled control siRNA (si-Control) knockdown. Data, mean ± SEM derived from three replicates. D, Nuclear RELB localization by confocal immunofluorescence microscopy following TRAF3-specific siRNA (siTRAF3b) or scrambled control siRNA (si-Control) knockdown at 24 hours. Cells were fixed and stained with antibodies against TRAF3, RELB, and Alexa fluor–conjugated secondary antibodies. Representative images show the localization of TRAF3 (green), RELB (purple), DAPI (blue), and merged images. Scale bar, 10 μm. E, NF-κB–mediated luciferase activity increases following TRAF3-specific siRNA (siTRAF3a-c). Renilla luciferase assay was performed on the same lysates and was used to adjust the differences in transfection efficiencies. All depicted samples were normalized to scrambled si-Control knockdown. Data, mean ± SEM derived from 6 replicates. Significant differences between samples and controls refer to *, P < 0.05; **, P < 0.01.

Close modal

TRAF3 inhibits aberrant and LTβ-enhanced NF-κB2/RELB protein, increases sensitivity to TNFα and cisplatin, as well as inhibits migration of HPV+ HNSCC cells

HPV+ HNSCCs display rapid growth and spread within the tonsils and regional lymph nodes, where ligands activating the alternative NF-κB pathway are expressed (4, 11). To investigate the upstream receptor-mediated activation of NF-κB in HPV+ lines, we first established if HPV+ and HPV HNSCC cell lines express mRNAs for the alternative pathway receptor lymphotoxin-beta receptor (LTβR), and two classic NF-κB pathway TNF receptors (TNFR1A and TNFR1B). We detected elevated mRNA expression of LTβR in HPV+ compared with HPV HNSCC cell lines (Fig. 5A, left, HOK cells as dotted line), while no significant difference was observed for either TNFRs (Fig. 5A, right). Next, we compared the functional effects of recombinant LTβ or TNFα protein on NF-κB reporter activity in HPV+ UMSCC47 cells and HPV UMSCC1 cells, establishing that LTβ as well as TNFα could induce NF-κB reporter activity in both (Fig. 5B; Supplementary Fig. S3A). We found that LTβ treatment attenuated TRAF3 and NF-κB2/p100 precursor protein, while enhancing RELB and processed NF-κB2/p52 protein levels (Fig. 5C; Supplementary Fig. S3B). Conversely, after knockdown of TRAF3, basal and LTβ-induced levels of RELB and NF-κB2/p52 proteins were substantially enhanced (Fig. 5C; Supplementary Fig. S3B), supporting the role of TRAF3 as a negative regulator of the alternative NF-κB pathway in HPV+ HNSCC cells.

Figure 5.

LTBR expression and LTβ-induced NF-κB activation in HPV+ HNSCC cell lines and enhancing effects of TRAF3 knockdown on alternative NF-κB pathway activation. A, mRNA-seq expression in eight cell lines, each with different HPV status were normalized to HOK cells (marked by dashed line). LTBR expression is relatively higher in HPV+ cell lines, while TNFR1A and TNFR1B exhibit no significant difference across HNSCC cell lines with different HPV status. The solid black line is the mean value. ****, P < 0.0001; ns, not significant, Student t test. B, NF-κB-RE reporter stably transfected UMSCC47 cells were plated at a cell density of 3,000/well in 96-well plate, cultured for 48 hours, then treated ± 10 ng/mL TNFα 16 hours or 50 ng/mL LTβ for 24 hours prior to GeneBLAzer Assay at 72 hours. Mean NF-κB activity normalized to no treatment ±SD for 6 replicates; P < 0.05 Student t test. C, UMSCC47 cells were transiently transfected with TRAF3 or control siRNA for 24 hours, followed by LTβ induction for 2 hours. The alternative NF-κB components RELB and NF-κB2 (p100 and p52) were examined in Western blot. β-Actin (Actin) was used as protein loading control. D, HPV+ UMSCC47 cells expressing TRAF3 were sensitized to TNFα and cisplatin at day 5 in the XTT assay. Cells were plated in 96-well plates, and the next day were treated with TNFα (25 ng/mL), LTβ (100 ng/mL), cisplatin (10 μmol/L) alone, or combination of TNFα, or LTβ with cisplatin. Cells transfected with control vector, blue; TRAF3 vector, red. Statistical significance (P < 0.05) was determined by the Student t test when compared with the control vector (*), untreated control (#), or cisplatin alone (+). The data are summarized from six replicates and are represented as mean ± SD. E, UMSCC47 cells were transiently transfected with siRNA targeting alternative pathway mediators IKKα and RELB, and cell proliferation was measured 3 days after transfection. Knockdown of IKKα or RELB decreased cell proliferation. All data are calculated relative to the samples transfected with control siRNA from 6 replicates and are represented as mean ± SD. *, P < 0.05; ***, P < 0.001; ****, P < 0.0001; Student t test. F, HPV+ UMSCC47 cells expressing TRAF3 or control vectors were plated in three wells each, and scratches were performed next day when the cells grew to confluency. Reduced cell migration via wound-healing assay was observed in TRAF3-transfected cells for 12 and 24 hours. D, The wound area percentage is shown for the control (blue) or TRAF3-expressing cells (red) at 12 and 24 hours after scratch. The cell migration assay was quantified from triplicates; *, P < 0.01 by Student t test.

Figure 5.

LTBR expression and LTβ-induced NF-κB activation in HPV+ HNSCC cell lines and enhancing effects of TRAF3 knockdown on alternative NF-κB pathway activation. A, mRNA-seq expression in eight cell lines, each with different HPV status were normalized to HOK cells (marked by dashed line). LTBR expression is relatively higher in HPV+ cell lines, while TNFR1A and TNFR1B exhibit no significant difference across HNSCC cell lines with different HPV status. The solid black line is the mean value. ****, P < 0.0001; ns, not significant, Student t test. B, NF-κB-RE reporter stably transfected UMSCC47 cells were plated at a cell density of 3,000/well in 96-well plate, cultured for 48 hours, then treated ± 10 ng/mL TNFα 16 hours or 50 ng/mL LTβ for 24 hours prior to GeneBLAzer Assay at 72 hours. Mean NF-κB activity normalized to no treatment ±SD for 6 replicates; P < 0.05 Student t test. C, UMSCC47 cells were transiently transfected with TRAF3 or control siRNA for 24 hours, followed by LTβ induction for 2 hours. The alternative NF-κB components RELB and NF-κB2 (p100 and p52) were examined in Western blot. β-Actin (Actin) was used as protein loading control. D, HPV+ UMSCC47 cells expressing TRAF3 were sensitized to TNFα and cisplatin at day 5 in the XTT assay. Cells were plated in 96-well plates, and the next day were treated with TNFα (25 ng/mL), LTβ (100 ng/mL), cisplatin (10 μmol/L) alone, or combination of TNFα, or LTβ with cisplatin. Cells transfected with control vector, blue; TRAF3 vector, red. Statistical significance (P < 0.05) was determined by the Student t test when compared with the control vector (*), untreated control (#), or cisplatin alone (+). The data are summarized from six replicates and are represented as mean ± SD. E, UMSCC47 cells were transiently transfected with siRNA targeting alternative pathway mediators IKKα and RELB, and cell proliferation was measured 3 days after transfection. Knockdown of IKKα or RELB decreased cell proliferation. All data are calculated relative to the samples transfected with control siRNA from 6 replicates and are represented as mean ± SD. *, P < 0.05; ***, P < 0.001; ****, P < 0.0001; Student t test. F, HPV+ UMSCC47 cells expressing TRAF3 or control vectors were plated in three wells each, and scratches were performed next day when the cells grew to confluency. Reduced cell migration via wound-healing assay was observed in TRAF3-transfected cells for 12 and 24 hours. D, The wound area percentage is shown for the control (blue) or TRAF3-expressing cells (red) at 12 and 24 hours after scratch. The cell migration assay was quantified from triplicates; *, P < 0.01 by Student t test.

Close modal

As we observed above that TRAF3 inhibited alternative NF-κB activation and cIAP1, which have been previously linked to proliferation, and TNFα and chemotherapy resistance (20), we evaluated if TRAF3 overexpression could inhibit tumor cell proliferation, or enhance the inhibitory effects of TNFα, LTβ, or chemotherapy agent cisplatin used for HNSCC. Transient TRAF3 expression resulted in a modest but significant decrease in cell proliferation and showed greater inhibitory effects in combination with TNFα, cisplatin, or both at day 3 (Supplementary Fig. S4A) and day 5 (Fig. 5D). In cell lines selected for TRAF3 and control vector expression, TRAF3 enhanced sensitivity of cells to the combination of TNFα and CDDP, but greater resistance and growth in response to TNFα alone (Supplementary Fig. S4B). While sensitization to TNFα and cisplatin was associated with TRAF3-inhibitory effects on alternative NF-κB components and cIAP1 above, we observed minimal modulation of BCL family members that contribute to survival (Supplementary Fig. S4C). To further examine the functional role of alternative NF-κB pathway, we knocked down IKKα and RELB to evaluate cell proliferation in HPV+ UMSCC47 cells. We observed a significant reduction in the cell proliferation after knockdown of either gene alone, with RELB knockdown having a greater effect than IKKα knockdown on cell proliferation (Fig. 5E). As HPV+ HNSCCs show increased propensity for malignant spread, we next performed a cell migration assay. We observed a significant delay in cell migration over a 24-hour time course in UMSCC47 cells expressing TRAF3 (Fig. 5F), before antiproliferative effects ∼10% of control are observed in XTT assay by day 3 (Supplementary Fig. S4A). Together, these data demonstrate that TRAF3 inhibits LTβ enhanced alternative pathway NF-κB2/p52 and RELB protein levels, increased sensitivity to TNFα and chemotheraputic agent as well as inhibits migration of HPV+ HNSCC cells.

Selection for TRAF3 expression or CRISPR knockout modulates colony formation and proliferation of HPV+ HNSCC

As TRAF3 appeared to reduce proliferation by 5 days following transient transfection, we enriched for UMSCC47 cells expressing TRAF3 or vector plasmid control by G418 selection, and examined the colony-forming capacity and proliferation of HPV+ UMSCC47 cells. We observed a significant reduction in colony formation of UMSCC47 cells that expressed increased TRAF3, compared with control (Fig. 6A and B). UMSCC47 cells selected for expression also demonstrated significantly reduced cell proliferation in TRAF3 versus vector control cells (Fig. 6C). Interestingly, we observed that TRAF3 expression declined with passage despite selection, consistent with the antiproliferative activity of a tumor suppressor (Supplementary Fig. S5). As the cell line UPCI-SCC-90 expressed higher endogenous levels of TRAF3 (Fig. 1E), we next examined effects of CRISPR knockout of TRAF3 on colony formation and cell proliferation. CRISPR depleted TRAF3 markedly enhanced colony formation and proliferation of UPCI-SCC-90 (Fig. 6D–F). Together, our data demonstrate that TRAF3 expression suppresses the proliferation of HPV+ HNSCC cells.

Figure 6.

TRAF3 expression or CRISPR knockout modulates colony formation and proliferation of HPV+ HNSCC. HPV+ UMSCC47 cells were transfected with His-TRAF3 or empty His-Control vector and selected by G418 for 2 weeks. A, The images of cell colony formation were taken using a microscope image system. B, Histogram showing the percentage of colonies produced with TRAF3-expressing cell line, compared with control vector. The colony numbers are represented as mean ± SD, derived from triplicates. *, P < 0.01 by Student t test. C, Cell proliferation of UMSCC47 cells selected for control or TRAF3 expression vector. Cells were plated in 96-well impedance plates and increasing cell densities were measured by impendence was determined at 40, 60, 80, and 100 hours. Mean cell density units +SD for 6 replicates. *, P < 0.05 Student t test for TRAF3 compared with control at each time point. D, Quantitative RT-PCR showing TRAF3 mRNA depletion in UPCI-SCC-90 CRISPR (Cr)TRAF3 knockout. E and F, Increased colony formation and cell proliferation of UPCI-SCC-90 CRISPR (Cr)TRAF3 knockout. Clonogenic survival was performed after plating 5,000 cells in 6-well plates. After 1 week (UPCI-SCC-90 CrTRAF3) or 2 weeks (UPCI-SCC-90 control), the colonies were fixed and stained with methylene blue in methanol. Colonies consisting of at least 30 cells were counted. Results from two independent experiments are presented. For proliferation, 1 × 105 UPCI-SCC-90 control or CrTRAF3 cells were plated in 12-well plates. At indicated time points, cells were collected and counted. Results from two independent experiments are presented.

Figure 6.

TRAF3 expression or CRISPR knockout modulates colony formation and proliferation of HPV+ HNSCC. HPV+ UMSCC47 cells were transfected with His-TRAF3 or empty His-Control vector and selected by G418 for 2 weeks. A, The images of cell colony formation were taken using a microscope image system. B, Histogram showing the percentage of colonies produced with TRAF3-expressing cell line, compared with control vector. The colony numbers are represented as mean ± SD, derived from triplicates. *, P < 0.01 by Student t test. C, Cell proliferation of UMSCC47 cells selected for control or TRAF3 expression vector. Cells were plated in 96-well impedance plates and increasing cell densities were measured by impendence was determined at 40, 60, 80, and 100 hours. Mean cell density units +SD for 6 replicates. *, P < 0.05 Student t test for TRAF3 compared with control at each time point. D, Quantitative RT-PCR showing TRAF3 mRNA depletion in UPCI-SCC-90 CRISPR (Cr)TRAF3 knockout. E and F, Increased colony formation and cell proliferation of UPCI-SCC-90 CRISPR (Cr)TRAF3 knockout. Clonogenic survival was performed after plating 5,000 cells in 6-well plates. After 1 week (UPCI-SCC-90 CrTRAF3) or 2 weeks (UPCI-SCC-90 control), the colonies were fixed and stained with methylene blue in methanol. Colonies consisting of at least 30 cells were counted. Results from two independent experiments are presented. For proliferation, 1 × 105 UPCI-SCC-90 control or CrTRAF3 cells were plated in 12-well plates. At indicated time points, cells were collected and counted. Results from two independent experiments are presented.

Close modal

TRAF3 expression enhances expression of and sensitizes HPV+ HNSCC to antiviral interferons

TRAF3 contributes to expression of type I IFN in response to other DNA virus infections in other tissues (13), but the expression of type-I IFN and relation to lower TRAF3 observed in HPV+HNSCC has not been examined. We analyzed RNA-seq data from our HNSCC cell lines and TCGA HNSCC tumors to compare the mRNA expression profiles for type-I interferon IFNA gene in HPV+ and HPV cell lines, as well as tumors. The HPV+ HNSCC cell lines displayed lower IFNA1 expression than normal HOK or HPV HNSCC cell lines (Fig. 7A, left). IFNA1 mRNA in HPV+ HNSCC tissues were also expressed at significantly lower levels when compared with HPV HNSCC tissues from TCGA data (Fig. 7A, right). To test the hypothesis that decreased TRAF3 may contribute to reduction in IFNA and be reversed by TRAF3, we compared IFNA1 expression in the two HPV+ HNSCC cell lines with decreased TRAF3 expression, UMSCC47 and UMSCC104, following transient transfection with control and TRAF3 vectors. TRAF3 induced a small but significant increase in the expression of IFNA1 in both cell lines (Fig. 7B). IFNA1 protein was also found to be modestly significantly increased in cell culture supernatants isolated from HPV+ UMSCC47 cells stably transfected with TRAF3 compared with control vector (Fig. 7C). UMSCC47 stably expressing increased TRAF3 and IFNA1 as above exhibited reduced proliferation and displayed enhanced sensitivity to exogenous IFNA1 or 2a (Fig. 7D). We explored if TRAF3 modulated mRNA expression of interferon response factors (IRF), which are transcription factors that mediate the transcription of type-I IFN (7, 11). However, we did not see consistent changes in IRF3, 5, or 7 mRNA expression in the HPV+ line selected for expression of the TRAF3 gene (Supplementary Fig. S6A). Further, while TRAF3-regulated IRF3 nuclear protein was also decreased in HPV+ UMSCC47 cells, the lack of nuclear IRF3 did not appear to be due to lower TRAF3 protein level alone, as expressing TRAF3 vector induced a minimal increase in expression of nuclear IRF3 in replicate experiments (Supplementary Fig. S6B). Moreover, even in HPV+ UMSCC47 with lower TRAF3 expression and low nuclear IRF3, nuclear IRF3 and type-I IFN remained inducible with a non-HPV RNA virus mimic and TLR3 ligand poly-I:C, unless TRAF3 was completely knocked out by CRISPR (Supplementary Fig. S7A–S7D). Conversely, UMSCC47 CRISPR/CAS9 TRAF3-9 KO rescued with TRAF3 vector showed no increase in baseline IRF3 or IFN, but did so following stimulation with poly-I:C (20 μg/mL; Supplementary Fig. S8A–S8D). Together, these findings are consistent with either a lack of strong activation of TRAF3-IRF3 signaling by integrated HPV in UMSCC47, or that IRF3 activation and type-I IFN expression may be further disrupted downstream of TRAF3 expression, as found in HPV E6/7-expressing keratinocytes in prior studies (6, 7).

Figure 7.

Expression of TRAF3 induces an antiviral interferon response, sensitizes to IFN, increases TP53 and RB protein expression, and modulates nuclear HPV E6 oncoprotein in HPV+ HNSCC cells. A, Antiviral cytokine IFNA1 mRNA expression in a panel of HPV+ vs. HPV HNSCC cell lines in this study (left) and HNSCC tissues from TCGA dataset (right). The mRNA expression for all samples is normalized to HOK cells (left) or normal tissues (right). P < 0.05 by Student t test: *, compared with normal tissues; #, comparison between HPV+ and normal controls. B and C, IFNA1 mRNA expression by qRT-PCR (B) and protein expression in supernatants by ELISA (C). Transient transfection of TRAF3 for 24 hours increased IFNA1 mRNA expression in HPV+ UMSCC47 and 104 cells and protein in supernatant of UMSCC47 cells. Data are derived from 6 replicates and represented as mean ± SD. *, P < 0.05; ns, not significant. D, UMSCC47 cells stably transfected with TRAF3 or control vector were plated overnight and treated with IFNA1 (5,000 U/ml) or IFNA2a (10,000 U/ml). Cell proliferation was measured by impedance. Data were calculated and are presented in bar graph as a mean of 6 replicates ± SD with Student t test. *, statistical significance (P < 0.05) when comparing IFNA1- or IFNA2a-treated cells with untreated cells. E, UMSCC47 selected to express TRAF3 show increased expression of tumor suppressor TP53 and RB. Expression of TRAF3, TP53, and RB is shown in HPV UMSCC46 cells as a positive control. β-Actin served as a loading control. F, UMSCC47 cells stably transfected with TRAF3 or control vector were plated overnight and transfected with siRNA targeting CDKN1A or control siRNA. Cell proliferation was measured by impedance. Data were calculated and are presented in bar graph as a mean of 6 replicates ± SD with Student t test. *, statistical significance (P < 0.05) when comparing cells transfected with CDKN1A siRNA with control siRNA. G, Cytoplasmic fractions (CF), nuclear fractions (NF), and whole cell lysates (WC) were isolated and HPV E6 protein was examined by Western blots. UMSCC47 cells selected to express TRAF3 show decreased E6 protein levels in nuclear lysates. H, Effects of E6 or TRAF3 overexpression on E6 protein in UMSCC47 cells. Left, E6 peptide-KLH protein–positive control for anti-E6 antibody. Top right, E6 and empty pcDNA vector were transiently transfected into UMSCC47 cells and effects in E6 protein expression were compared. A slight increase in E6 protein was detected. Bottom right, A slight decrease in E6 expression was detected 72 hours after TRAF3 expression. Actin was used as the loading control.

Figure 7.

Expression of TRAF3 induces an antiviral interferon response, sensitizes to IFN, increases TP53 and RB protein expression, and modulates nuclear HPV E6 oncoprotein in HPV+ HNSCC cells. A, Antiviral cytokine IFNA1 mRNA expression in a panel of HPV+ vs. HPV HNSCC cell lines in this study (left) and HNSCC tissues from TCGA dataset (right). The mRNA expression for all samples is normalized to HOK cells (left) or normal tissues (right). P < 0.05 by Student t test: *, compared with normal tissues; #, comparison between HPV+ and normal controls. B and C, IFNA1 mRNA expression by qRT-PCR (B) and protein expression in supernatants by ELISA (C). Transient transfection of TRAF3 for 24 hours increased IFNA1 mRNA expression in HPV+ UMSCC47 and 104 cells and protein in supernatant of UMSCC47 cells. Data are derived from 6 replicates and represented as mean ± SD. *, P < 0.05; ns, not significant. D, UMSCC47 cells stably transfected with TRAF3 or control vector were plated overnight and treated with IFNA1 (5,000 U/ml) or IFNA2a (10,000 U/ml). Cell proliferation was measured by impedance. Data were calculated and are presented in bar graph as a mean of 6 replicates ± SD with Student t test. *, statistical significance (P < 0.05) when comparing IFNA1- or IFNA2a-treated cells with untreated cells. E, UMSCC47 selected to express TRAF3 show increased expression of tumor suppressor TP53 and RB. Expression of TRAF3, TP53, and RB is shown in HPV UMSCC46 cells as a positive control. β-Actin served as a loading control. F, UMSCC47 cells stably transfected with TRAF3 or control vector were plated overnight and transfected with siRNA targeting CDKN1A or control siRNA. Cell proliferation was measured by impedance. Data were calculated and are presented in bar graph as a mean of 6 replicates ± SD with Student t test. *, statistical significance (P < 0.05) when comparing cells transfected with CDKN1A siRNA with control siRNA. G, Cytoplasmic fractions (CF), nuclear fractions (NF), and whole cell lysates (WC) were isolated and HPV E6 protein was examined by Western blots. UMSCC47 cells selected to express TRAF3 show decreased E6 protein levels in nuclear lysates. H, Effects of E6 or TRAF3 overexpression on E6 protein in UMSCC47 cells. Left, E6 peptide-KLH protein–positive control for anti-E6 antibody. Top right, E6 and empty pcDNA vector were transiently transfected into UMSCC47 cells and effects in E6 protein expression were compared. A slight increase in E6 protein was detected. Bottom right, A slight decrease in E6 expression was detected 72 hours after TRAF3 expression. Actin was used as the loading control.

Close modal

TRAF3 expression enhances TP53 and RB tumor suppressor and inhibits E6 protein expression

As repression of tumor suppressor proteins TP53 and RB by HPV contributes to the proliferation of HPV+ cancers (21, 22), we examined if TRAF3 may modulate expression of TP53 and RB. Compared with the HPV mtTP53 line UMSCC46, UMSCC47 expressed low basal levels of TP53 and RB protein expression (Fig. 7E). Unexpectedly, TP53 and RB expression was reproducibly enhanced in UMSCC47 cells selected for expression of TRAF3 or after transient transfection (Fig. 7E; Supplementary Fig. S9A). UMSCC104 cells with the lowest TRAF3 expression also showed enhanced TP53 and RB expression after transient transfection with TRAF3 (Supplementary Fig. S9B). In UMSCC47 TRAF3-expressing cells, the increase in TP53 was not attributable to increased mRNA, while RB mRNA was increased ∼2-fold as measured by qRT-PCR (Supplementary Fig. S9C). TP53 siRNA knockdown attenuated the antiproliferative effects of TRAF3 (Supplementary Fig. S7D). The increase in TP53 was associated with enhanced expression of TP53-regulated cyclin-dependent kinase CDKN1A(p21), and proliferation was enhanced by siRNA knockdown of CDKN1A(p21) in UMSCC47 expressing TRAF3 (Fig. 7F and Supplementary Fig. S9E), supporting a contribution of this cell-cycle regulator to the antiproliferative effects of TRAF3 and TP53.

As HPV E6 oncoprotein mediates suppression of TP53 (22), we explored whether TRAF3 may affect HPV E6 levels protein in UMSCC47 cells selected to express TRAF3 or control vector. HPV E6 protein localized predominantly in the nucleus in the control cell line and was partially decreased in the TRAF3-expressing UMSCC47 cell line (Fig. 7G). We observed minimal change in transcription of E6 mRNA (Supplementary Fig. S10A). However, we observed only a minimal increase in E6 protein after transfection with an E6 expression vector that increased mRNA ∼400-fold (Fig. 7H; Supplementary Fig. S10B), indicating that the very low level of E6 detected is not primarily determined by transcriptional regulation (Fig. 7H). Conversely, we also observed only a minimal decrease in E6 protein at 72 hours after transient transfection with TRAF3 (Fig. 7H) and did not observe higher or lower MW bands, or enhancement by proteasome inhibitors, that would support a ubiquitination or proteasome-dependent mechanism (Supplementary Fig. S10C). Together, these observations suggest that the increase in TP53 with TRAF3 within 24 hours after transfection is likely dependent on mechanisms other than modulation of E6 protein expression alone.

TRAF3 copy loss, mutation, and/or decreased expression was significantly enriched in a subset of HPV+ HNSCC tumors (10), suggesting that TRAF3 could be a novel suppressor of HPV viral and tumor pathogenesis. Previously, TRAF3 deficiency was implicated in impaired Toll-like receptor 3 responses and increased susceptibility to herpes simplex virus (HSV)–induced encephalitis (23), hematologic malignancies, and Epstein–Barr virus (EBV)–related nasopharyngeal cancers, but not HPV cancers. We observed that oropharyngeal HNSCC linked to HPV exhibit aberrant nuclear activation of the alternative NF-κB pathway (24), but the significance and function and role of deficient TRAF3 in alternative NF-κB activation have not been investigated previously in HPV+ HNSCC cell lines and tissues. Here, we demonstrate for the first time that decreased TRAF3 expression is linked to aberrant expression and activation of alternative NF-κB2/RELB transcription factors, contributes to reduced expression of antiviral type-I IFN and tumor suppressor TP53 and RB proteins, which are deregulated in HPV+ HNSCC (Supplementary Fig. S11). Supporting this role, ectopic TRAF3 expression in HPV+ HNSCC with decreased endogenous TRAF3 inversely modulated these key targets and suppressed several hallmarks of cancer, including cell colony formation, proliferation, and migration, while sensitizing HPV+ HNSCC to immune IFNs, TNFα, and chemotherapy agent cisplatin. Complementing our findings, one of our laboratories currently obtained evidence that deletions or mutations in TRAF3 in HPV+ HNSCC tumors are associated with increased expression of an NF-κB transcriptional program, and episomal HPV infection (25). Interestingly, TRAF3-deficient HPV+ HNSCC also appear to be linked to a better prognosis (25), than HPV+ or HPV tumors with PIK3CA, ΔNp63, FADD, cIAP1/2, and other genomic alterations (i.e., TP53; ref. 10), implicated in modulation of NF-κB or other pathways important in pathogenicity (26–29). The findings in the present study support the addition of TRAF3 as a tumor suppressor in HPV+ HNSCC.

The recent discovery of deep deletions on chromosome 14q32.32 containing the TRAF3 coding region and 3 deleterious loss-of-function mutations by TCGA project was previously unrecognized in HPV+ HNSCC (10). Here, we further defined an increased frequency of hetero- and homozygous deletions of TRAF3 in HPV+ HNSCC, in contrast to HPV tumors, which often display copy gain. Furthermore, we observed that TRAF3 genetic alterations and expression significantly correlated with expression of alternative NF-κB pathway components in HNSCC tissues. TRAF3 gene copy loss, inactivating mutations, and decreased expression of TRAF3 have been previously observed only in non-HPV cancers. These include multiple myeloma (MM; ref. 16), B-cell lymphomas (30), and EBV–infected human nasopharyngeal carcinomas (NPC; ref. 31). Annunziata and colleagues found that 4.4% of 451 patients with MM had TRAF3 genetic defects, such as silencing, homozygous deletion, or somatic mutation, and 17 MM cases exhibited TRAF3 mRNA expression 13-fold below the median level of the cohort, predominantly in association with these genetic alterations (16). Chung and colleagues detected deletion or missense mutations of TRAF3, TRAF2, and A20 in 3 of 33 (9.1%) of primary NPC tumor specimens (31). Together, these observations in different cancer types support the potential relevance of genetic alterations in HPV+ HNSCC and suggest that decreased TRAF3 facilitates the pathogenesis of tumors within the hematopoietic or lymphatic microenvironment.

We previously reported that alternative pathway NF-κB2 and RELB subunits are often aberrantly expressed, and localized to the nucleus in oropharyngeal HNSCC tumors linked to HPV (24), but the possible relationship to genomic alterations, other factors and to the malignant phenotype was unknown. Here, our analysis of TCGA HNSCC data indicated that TRAF3 deletion is associated with increased mRNA expression of molecules involved in the alternative NF-κB pathway, such as NIK, cIAPs, RELB, and NF-κB2. Co-occurrence and mutual exclusivity analyses for key components of the NF-κB pathways revealed a significant association of altered TRAF3 with molecules involved in an alternative NF-κB pathway in HPV+, but not in HPV HNSCC tissues. We further demonstrated that HPV+ HNSCC cell lines with decreased endogenous TRAF3 also display aberrant expression and nuclear activation of the alternative NF-κB pathway. Furthermore, this potential of decreased TRAF3 to foster expression and activation of alternative NF-κB in HPV+ HNSCC tumors could potentially be enhanced within the local tonsillar and lymphatic microenvironment, where alternative NF-κB signal inducers such as LTβ are abundant (32).

LTβ is a key signaling ligand that binds to surface receptor LTβR and activates the alternative NF-κB pathway (33). We originally detected aberrant activation of the alternative pathway in HPV HNSCC and demonstrated that activation could be enhanced by LTβ stimulation (34). Here, we observed increased LTβR expression compared with HPV cell lines and showed that LTβ could further enhance NF-κB activation in HPV+ HNSCC cells expressing lower endogenous TRAF3. TRAF3 knockdown further enhanced aberrant and LTβ-stimulated expression of the alternative NF-κB2 subunit p52 and RELB. Interestingly, in a subset of EBV-infected nasopharyngeal HNSCC that harbor TRAF3 alterations, Or and colleagues demonstrated 12p13.3 copy-number gains and increased expression of LTβR (35). Overexpression of LTβR in nasopharyngeal epithelial cells resulted in increased NF-κB activity and cell proliferation (36). Together, these data implicate TRAF3 and/or LTβR genomic alterations, along with HPV E6 and EBV LMP1 viral oncogenes that promote alternative NF-κB pathway activation, in viral pathogenesis of oro- and nasopharyngeal HNSCC. LMP-1 has been shown to bind TRAF3 and enhance NIK-dependent alternative pathway activation (36). HPV E6 is reported to promote alternative NF-κB activation via a mechanism requiring its PDZ domain implicated in binding phosphatases (8). Our data support a role for deficient TRAF3 expression in enhancing NIK, cIAP1, IKKα, NF-κB2-p52, and RELB proteins, but we have found it exceedingly difficult to find conditions to detect HPV16 E6 protein and an association with TRAF3 in HNSCC.

TRAF3 has been reported to have a unique dual role in positively regulating type-I IFNs in response to different DNA viruses in other tissues (11). IFNA1 potentially has antiviral effects through inhibition of viral protein expression, proliferation, as well as immunostimulatory properties, that link innate and adaptive immunity (37). In this study, we observed significantly lower expression of IFNA1 in HPV+ HNSCC lines, consistent with the lower expression of IFNA1 detected in HPV+ compared with HPV tumor tissues from TCGA data. Previous studies have shown that HPV infection can also induce ubiquitin carboxyl-terminal hydrolase L1 (UCHL1), which could inhibit TRAF3 K63 ubiquination, important in downstream TBK-IRF3–mediated type-I IFN expression (7). In addition, we have shown a significant increase in both type-I IFN mRNA and protein expression by overexpressing TRAF3 in HPV+ UMSCC cell lines with low endogenous levels or TRAF3 rescue in TRAF3-KO cell lines. TRAF3 also sensitized HPV+ cells to antiproliferative effects of exogenous IFNA1 that may be produced by cells in the tumor microenvironment, or IFN2A, which is used clinically.

A novel finding of this study supporting the importance of decreased TRAF3 in viral pathogenesis is that overexpression of TRAF3 enhanced the expression of TP53 and RB, which are critical tumor suppressor proteins inactivated by HPV E6 and E7 oncoproteins (21, 22, 38, 39). Proliferation was enhanced by knockdown of TP53 and TP53-regulated CDKN1A(p21) in cells selected to express TRAF3, but not in control cells. However, the exact mechanism by which TRAF3 modulates these tumor suppressors and the relationship of these effects to E6 and E7 is complicated by the difficulty in detecting these proteins. The minor reduction in E6 observed with TRAF3 was not accompanied by appearance of increased or decreased MW bands, and/or modulated by proteasome inhibitors, that would support a ubiquitin or proteasome-dependent mechanism (38). Further, the effects of TRAF3 on decreased E6 were detected at later time points, compared with effects on TP53 and RB expression or NF-κB activation. The different kinetics suggest that TRAF3 modulates E6, TP53, and RB expression by different transcriptional or posttranslational mechanisms.

Of potential therapeutic relevance, TRAF3 re-expression in HPV+ HNSCC cells with low endogenous expression sensitized them to inhibition by cisplatin chemotherapy and TNFα, an important cell death ligand induced by radiation and immune therapy (40, 41). Cisplatin is the most active chemotherapeutic agent in HNSCC (42), leading to DNA damage and subsequently promoting cell death (43). TNFα is known to bind to TNF receptors and induce apoptotic cell death in normal cells, but HNSCC are relatively resistant to TNFα (28, 44). NF-κB and cIAPs have been implicated in resistance to TNFα and chemotherapy (20, 45). Our data suggest that TRAF3 expression can inhibit cIAP1 expression and activation of the alternative NF-κB pathway, and sensitize HPV+ HNSCC to TNFα and cisplatin-induced cell death. We have recently reported that IAP1 inhibitors can sensitize HPV HNSCC with gene copy gain and expression of IAP1 to TNF, chemotherapy, and radiotherapy (28, 46, 47), supporting future investigation of these agents in HPV+ HNSCCs expressing deficient TRAF3 and/or cIAP1.

Although both HPV+ HNSCCs and NPCs develop within the adenotonsillar tissue and exhibit high rates of spread to regional lymph nodes, the mechanisms for this predilection are not well understood. Intriguingly, TRAF3 re-expression inhibited cell migration in vitro, suggesting that deficient TRAF3 could contribute to enhanced migration, underlying the high rates of regional spread of both HPV+ HNSCC and NPC in vivo. The alternative of NF-κB pathway could have a broader role in migration in HNSCC, as we found that IKKα and RELB knockdown decreased cell migration and proliferation in HPV HNSCC (34, 48). Together, the unique genomic alterations in TRAF3 and aberrantly activated alternative NF-κB pathway components such as cIAPs or NIK, could serve as potential markers and targets for novel therapies of HPV+ HNSCC.

C. Van Waes reports receiving a commercial research grant from Astex Pharamaceuticals. No potential conflicts of interest were disclosed by the other authors.

Conception and design: J. Zhang, C.M. Annunziata, Z. Chen, C. Van Waes

Development of methodology: J. Zhang, X. Yang, P.E. Clavijo, Z. Chen

Acquisition of data (provided animals, acquired and managed patients, provided facilities, etc.): J. Zhang, T. Chen, X. Yang, S.S. Späth, C. Silvin, N. Issaeva, Z. Chen

Analysis and interpretation of data (e.g., statistical analysis, biostatistics, computational analysis): J. Zhang, T. Chen, X. Yang, H. Cheng, S.S. Späth, P.E. Clavijo, J. Chen, N. Issaeva, W.G. Yarbrough, Z. Chen, C. Van Waes

Writing, review, and/or revision of the manuscript: J. Zhang, T. Chen, S.S. Späth, P.E. Clavijo, J. Chen, W.G. Yarbrough, C.M. Annunziata, Z. Chen, C. Van Waes

Administrative, technical, or material support (i.e., reporting or organizing data, constructing databases): X. Yang, Z. Chen

Study supervision: Z. Chen, C. Van Waes

We thank Dr. Cheng-Ming Chiang and S.-Y. Wu (UT Southwestern Medical Center) for providing HPV16E6 expression plasmid and their helpful suggestions. We thank Drs. Liu Yang (University of Maryland School of Medicine) and WanJun Chen (NIDCR/NIH) for reading the manuscript and comments.

This work was supported by the Intramural Research Program of The National Institute on Deafness and Other Communication Disorders. J. Zhang, T. Chen, H. Cheng, X. Yang, P. Clavijo, J. Coupar, C. Silvin, Z. Chen, and C. Van Waes are supported by NIDCD intramural projects ZIA-DC-000016, 73, 74. C.M. Annunziata is supported by the NCI.

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.

1.
Siegel
RL
,
Miller
KD
,
Jemal
A
. 
Cancer statistics
,
2016. CA Cancer J Clin
2016
;
66
:
7
30
.
2.
Leemans
CR
,
Braakhuis
BJ
,
Brakenhoff
RH
. 
The molecular biology of head and neck cancer
.
Nat Rev Cancer
2011
;
11
:
9
22
.
3.
Ang
KK
,
Harris
J
,
Wheeler
R
,
Weber
R
,
Rosenthal
DI
,
Nguyen-Tan
PF
, et al
Human papillomavirus and survival of patients with oropharyngeal cancer
.
N Engl J Med
2010
;
363
:
24
35
.
4.
Gillison
ML
,
Koch
WM
,
Capone
RB
,
Spafford
M
,
Westra
WH
,
Wu
L
, et al
Evidence for a causal association between human papillomavirus and a subset of head and neck cancers
.
J Natl Cancer I
2000
;
92
:
709
20
.
5.
Chaturvedi
AK
,
Engels
EA
,
Pfeiffer
RM
,
Hernandez
BY
,
Xiao
WH
,
Kim
E
, et al
Human papillomavirus and rising oropharyngeal cancer incidence in the United States
.
J Clin Oncol
2011
;
29
:
4294
301
.
6.
Nees
M
,
Geoghegan
JM
,
Hyman
T
,
Frank
S
,
Miller
L
,
Woodworth
CD
. 
Papillomavirus type 16 oncogenes downregulate expression of interferon-responsive genes and upregulate proliferation-associated and NF-kappaB-responsive genes in cervical keratinocytes
.
J Virol
2001
;
75
:
4283
96
.
7.
Karim
R
,
Tummers
B
,
Meyers
C
,
Biryukov
JL
,
Alam
S
,
Backendorf
C
, et al
Human papillomavirus (HPV) upregulates the cellular deubiquitinase UCHL1 to suppress the keratinocyte's innate immune response
.
PLoS Pathog
2013
;
9
:
e1003384
.
8.
James
MA
,
Lee
JH
,
Klingelhutz
AJ
. 
Human papillomavirus type 16 E6 activates NF-kappaB, induces cIAP-2 expression, and protects against apoptosis in a PDZ binding motif-dependent manner
.
J Virol
2006
;
80
:
5301
7
.
9.
Schiffman
M
,
Doorbar
J
,
Wentzensen
N
,
de Sanjose
S
,
Fakhry
C
,
Monk
BJ
, et al
Carcinogenic human papillomavirus infection
.
Nat Rev Dis Primers
2016
;
2
:
16086
.
10.
Comprehensive genomic characterization of head and neck squamous cell carcinomas
.
Nature
2015
;
517
:
576
82
.
11.
Häcker H1
TP
,
Karin
M
. 
Expanding TRAF function: TRAF3 as a tri-faced immune regulator
.
Nat Rev Immunol
2011
;
11
:
457
68
.
12.
Annunziata
CM
,
Davis
RE
,
Demchenko
Y
,
Bellamy
W
,
Gabrea
A
,
Zhan
F
, et al
Frequent engagement of the classical and alternative NF-kappaB pathways by diverse genetic abnormalities in multiple myeloma
.
Cancer Cell
2007
;
12
:
115
30
.
13.
Oganesyan
G
,
Saha
SK
,
Guo
B
,
He
JQ
,
Shahangian
A
,
Zarnegar
B
, et al
Critical role of TRAF3 in the Toll-like receptor-dependent and -independent antiviral response
.
Nature
2006
;
439
:
208
11
.
14.
Tang
AL
,
Owen
JH
,
Hauff
SJ
,
Park
JJ
,
Papagerakis
S
,
Bradford
CR
, et al
Head and neck cancer stem cells: the effect of HPV—an in vitro and mouse study
.
Otolaryngol Head Neck Surg
2013
;
149
:
252
60
.
15.
Tang
AL
,
Hauff
SJ
,
Owen
JH
,
Graham
MP
,
Czerwinski
MJ
,
Park
JJ
, et al
UM-SCC-104: a new human papillomavirus-16-positive cancer stem cell-containing head and neck squamous cell carcinoma cell line
.
Head Neck
2012
;
34
:
1480
91
.
16.
Brenner
JC
,
Graham
MP
,
Kumar
B
,
Saunders
LM
,
Kupfer
R
,
Lyons
RH
, et al
Genotyping of 73 UM-SCC head and neck squamous cell carcinoma cell lines
.
Head Neck
2010
;
32
:
417
26
.
17.
Franken
NA
,
Rodermond
HM
,
Stap
J
,
Haveman
J
,
van Bree
C
. 
Clonogenic assay of cells in vitro
.
Nat Protoc
2006
;
1
:
2315
9
.
18.
Oeckinghaus
A
,
Ghosh
S
. 
The NF-kappaB family of transcription factors and its regulation
.
Cold Spring Harb Perspect Biol
2009
;
1
:
a000034
.
19.
Havard
L
,
Rahmouni
S
,
Boniver
J
,
Delvenne
P
. 
High levels of p105 (NFKB1) and p100 (NFKB2) proteins in HPV16-transformed keratinocytes: role of E6 and E7 oncoproteins
.
Virology
2005
;
331
:
357
66
.
20.
Wang
CY
,
Mayo
MW
,
Korneluk
RG
,
Goeddel
DV
,
Baldwin
AS
 Jr
. 
NF-kappaB antiapoptosis: induction of TRAF1 and TRAF2 and c-IAP1 and c-IAP2 to suppress caspase-8 activation
.
Science
1998
;
281
:
1680
3
.
21.
Yim
EK
,
Park
JS
. 
The role of HPV E6 and E7 oncoproteins in HPV-associated cervical carcinogenesis
.
Cancer Res Treat
2005
;
37
:
319
24
.
22.
Thomas
MC
,
Chiang
CM
. 
E6 oncoprotein represses p53-dependent gene activation via inhibition of protein acetylation independently of inducing p53 degradation
.
Mol Cell
2005
;
17
:
251
64
.
23.
Perez de Diego
R
,
Sancho-Shimizu
V
,
Lorenzo
L
,
Puel
A
,
Plancoulaine
S
,
Picard
C
, et al
Human TRAF3 adaptor molecule deficiency leads to impaired Toll-like receptor 3 response and susceptibility to herpes simplex encephalitis
.
Immunity
2010
;
33
:
400
11
.
24.
Allen
C
,
Saigal
K
,
Nottingham
L
,
Arun
P
,
Chen
Z
,
Van Waes
C
. 
Bortezomib-induced apoptosis with limited clinical response is accompanied by inhibition of canonical but not alternative nuclear factor-kappa B subunits in head and neck cancer
.
Clinical Cancer Research
2008
;
14
:
4175
85
.
25.
Hajek
M
,
Sewell
A
,
Kaech
S
,
Burtness
B
,
Yarbrough
WG
,
Issaeva
N
. 
TRAF3/CYLD mutations identify a distinct subset of human papillomavirus-associated head and neck squamous cell carcinoma
.
Cancer-Am Cancer Soc
2017
;
123
:
1778
90
.
26.
Mohan
S
,
Vander Broek
R
,
Shah
S
,
Eytan
DF
,
Pierce
ML
,
Carlson
SG
, et al
MEK inhibitor PD-0325901 overcomes resistance to PI3K/mTOR inhibitor PF-5212384 and potentiates antitumor effects in human head and neck squamous cell carcinoma
.
Clin Cancer Res
2015
;
21
:
3946
56
.
27.
Yang
X
,
Lu
H
,
Yan
B
,
Romano
RA
,
Bian
Y
,
Friedman
J
, et al
ΔNp63 versatilely regulates a broad NF-κB gene program and promotes squamous epithelial proliferation, migration, and inflammation
.
Cancer Res
2011
;
71
:
3688
700
.
28.
Eytan
DF
,
Snow
GE
,
Carlson
S
,
Derakhshan
A
,
Saleh
A
,
Schiltz
S
, et al
SMAC mimetic birinapant plus radiation eradicates human head and neck cancers with genomic amplifications of cell death genes FADD and BIRC2
.
Cancer Res
2016
;
76
:
5442
54
.
29.
Lee
TL
,
Yeh
J
,
Friedman
J
,
Yan
B
,
Yang
X
,
Yeh
NT
, et al
A signal network involving coactivated NF-kappaB and STAT3 and altered p53 modulates BAX/BCL-XL expression and promotes cell survival of head and neck squamous cell carcinomas
.
Int J Cancer
2008
;
122
:
1987
98
.
30.
Mambetsariev
N
,
Lin
WW
,
Stunz
LL
,
Hanson
BM
,
Hildebrand
JM
,
Bishop
GA
. 
Nuclear TRAF3 is a negative regulator of CREB in B cells
.
Proc Natl Acad Sci U S A
2016
;
113
:
1032
37
.
31.
Chung
GT
,
Lou
WP
,
Chow
C
,
To
KF
,
Choy
KW
,
Leung
AW
, et al
Constitutive activation of distinct NF-kappaB signals in EBV-associated nasopharyngeal carcinoma
.
J Pathol
2013
;
231
:
311
22
.
32.
Ganeff
C
,
Remouchamps
C
,
Boutaffala
L
,
Benezech
C
,
Galopin
G
,
Vandepaer
S
, et al
Induction of the alternative NF-kappaB pathway by lymphotoxin alphabeta (LTalphabeta) relies on internalization of LTbeta receptor
.
Mol Cell Biol
2011
;
31
:
4319
34
.
33.
Sun
SC
. 
Non-canonical NF-kappaB signaling pathway
.
Cell Res
2011
;
21
:
71
85
.
34.
Nottingham
LK
,
Yan
CH
,
Yang
X
,
Si
H
,
Coupar
J
,
Bian
Y
, et al
Aberrant IKKalpha and IKKbeta cooperatively activate NF-kappaB and induce EGFR/AP1 signaling to promote survival and migration of head and neck cancer
.
Oncogene
2014
;
33
:
1135
47
.
35.
Or
YY
,
Chung
GT
,
To
KF
,
Chow
C
,
Choy
KW
,
Tong
CY
, et al
Identification of a novel 12p13.3 amplicon in nasopharyngeal carcinoma
.
J Pathol
2010
;
220
:
97
107
.
36.
Xie
P
,
Bishop
GA
. 
Roles of TNF receptor-associated factor 3 in signaling to B lymphocytes by carboxyl-terminal activating regions 1 and 2 of the EBV-encoded oncoprotein latent membrane protein
.
J Immunol
2004
;
173
:
5546
55
.
37.
Ivashkiv
LB
,
Donlin
LT
. 
Regulation of type I interferon responses
.
Nat Rev Immunol
2014
;
14
:
36
49
.
38.
Huibregtse
JM
,
Scheffner
M
,
Howley
PM
. 
Localization of the E6-Ap regions that direct human papillomavirus E6 binding, association with P53, and ubiquitination of associated proteins
.
Mol Cell Biol
1993
;
13
:
4918
27
.
39.
Boyer
SN
,
Wazer
DE
,
Band
V
. 
E7 protein of human papilloma virus-16 induces degradation of retinoblastoma protein through the ubiquitin-proteasome pathway
.
Cancer Res
1996
;
56
:
4620
24
.
40.
Hallahan
DE
,
Spriggs
DR
,
Beckett
MA
,
Kufe
DW
,
Weichselbaum
RR
. 
Increased tumor necrosis factor alpha mRNA after cellular exposure to ionizing radiation
.
Proc Natl Acad Sci U S A
1989
;
86
:
10104
7
.
41.
Zhang
M
,
Kong
F
,
Qian
J
,
Chen
M
,
Lawrence
TS
. 
Radiation-induced lung apoptosis is mediated by TNF-alpha action
.
Int J Radiat Oncol
2005
;
63
:
S83
S84
.
42.
Seiwert
TY
,
Salama
JK
,
Vokes
EE
. 
The chemoradiation paradigm in head and neck cancer
.
Nat Clin Pract Oncol
2007
;
4
:
156
71
.
43.
Ummat
A
,
Rechkoblit
O
,
Jain
R
,
Roy Choudhury
J
,
Johnson
RE
,
Silverstein
TD
, et al
Structural basis for cisplatin DNA damage tolerance by human polymerase eta during cancer chemotherapy
.
Nat Struct Mol Biol
2012
;
19
:
628
32
.
44.
Duffey DC1
C-BC
,
Chen
Z
,
Ondrey
FG
,
Nejad-Sattari
M
,
Dong
G
,
Van Waes
C
. I 
nhibition of transcription factor nuclear factor-kappaB by a mutant inhibitor-kappaBalpha attenuates resistance of human head and neck squamous cell carcinoma to TNF-alpha caspase-mediated cell death
.
Br J Cancer
2000
;
83
:
1367
74
.
45.
Tse
BW
,
Scott
KF
,
Russell
PJ
. 
Paradoxical roles of tumour necrosis factor-alpha in prostate cancer biology
.
Prostate Cancer
2012
;
2012
:
128965
.
46.
Eytan
DF
,
Snow
GE
,
Carlson
SG
,
Schiltz
S
,
Chen
Z
,
Van Waes
C
. 
Combination effects of SMAC mimetic birinapant with TNFalpha, TRAIL, and docetaxel in preclinical models of HNSCC
.
Laryngoscope
2015
;
125
:
E118
24
.
47.
Xiao
R
,
An
Y
,
Derakhshan
A
,
Chen
Z
,
Schmitt
N
,
Van Waes
C
. 
Novel dual cIAP1/XIAP antagonist ASTX660 activity in preclinical models of human papillomavirus(+) and (-) head and neck squamous cell carcinoma
.
Amer Assn Cancer Res
2017
;
abstr 1086
.
48.
Das R
CJ
,
Saleh
A
,
Clavijo
PE
,
Chen
Z
,
Van Waes
C
. 
LTβ/LTβ receptor mediates alternative NF-κB activation through NIK and RELB/NF-kB2 to promote cell migration of HNSCC
.
Proc Amer Assn Cancer Res
2017
;
abstr 358
.

Supplementary data